1
|
Shelford J, Burgess SG, Rostkova E, Richards MW, Larocque G, Sampson J, Tiede C, Fielding AJ, Daviter T, Tomlinson DC, Calabrese AN, Pfuhl M, Bayliss R, Royle SJ. Structural characterization and inhibition of the interaction between ch-TOG and TACC3. J Cell Biol 2025; 224:e202407002. [PMID: 40105698 PMCID: PMC11921806 DOI: 10.1083/jcb.202407002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/19/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
The mitotic spindle is a bipolar array of microtubules, radiating from the poles which each contain a centrosome, embedded in pericentriolar material. Two proteins, ch-TOG and TACC3, have multiple functions at the mitotic spindle due to operating either alone, together, or in complex with other proteins. To distinguish these activities, we need new molecular tools to dissect their function. Here, we present the structure of the α-helical bundle domain of ch-TOG that mediates its interaction with TACC3 and a structural model describing the interaction, supported by biophysical and biochemical data. We have isolated Affimer tools to precisely target the ch-TOG-binding site on TACC3 in live cells, which displace ch-TOG without affecting the spindle localization of other protein complex components. Inhibition of the TACC3-ch-TOG interaction led unexpectedly to fragmentation of the pericentriolar material in metaphase cells and delayed mitotic progression, uncovering a novel role of TACC3-ch-TOG in maintaining pericentriolar material integrity during mitosis to ensure timely cell division.
Collapse
Affiliation(s)
- James Shelford
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Coventry, UK
| | - Selena G. Burgess
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Elena Rostkova
- School of Cardiovascular and Metabolic Medicine and Sciences and Randall Centre, King’s College London, Guy’s Campus, London, UK
| | - Mark W. Richards
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Gabrielle Larocque
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Coventry, UK
| | - Josephina Sampson
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Christian Tiede
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Alistair J. Fielding
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Tina Daviter
- Institute of Cancer Research, Chester Beatty Laboratories, London, UK
| | - Darren C. Tomlinson
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Antonio N. Calabrese
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Mark Pfuhl
- School of Cardiovascular and Metabolic Medicine and Sciences and Randall Centre, King’s College London, Guy’s Campus, London, UK
| | - Richard Bayliss
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Stephen J. Royle
- Centre for Mechanochemical Cell Biology, Warwick Medical School, University of Warwick, Coventry, UK
| |
Collapse
|
2
|
Lengerli D, Çalışkan ÖA, Çalışkan K, Saatci Ö, Lim C, Vempati S, Çalışkan B, Şahin Ö, Banoglu E. Isoxazole-pyrimidine derivatives as TACC3 inhibitors: A novel modality to targeted cancer therapy. Bioorg Chem 2025; 156:108204. [PMID: 39889548 DOI: 10.1016/j.bioorg.2025.108204] [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: 12/10/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025]
Abstract
Inhibiting the function of transforming acidic coiled-coil 3 (TACC3) offers a promising therapeutic approach for various cancers, such as breast, ovarian, and lung cancers.Our previous work introduced BO-264 as a novel chemotype for inhibiting TACC3 function, though it exhibited relatively low metabolic stability. In this study, sixty-two compounds were designed and synthesized to modify the structure of BO-264 to improve its metabolic stability while maintaining its potency. The tractable SAR results obtained by these novel analogs indicated that appropriate substitutions on the left-end phenyl-isoxazole and right-end morpholine groups improved metabolic stability while preserving potency. Among these, compound 13b exhibited approximately sevenfold improvement in metabolic stability and bioavailability while maintaining strong potency and a favorable safety profile. 13b markedly increased the levels of p-Histone H3 (Ser10), cleaved PARP, and p-H2AX (Ser139), indicative of mitotic arrest, apoptosis, and DNA damage, respectively. In addition, the protein-drug binding assay, DARTS, identified TACC3 as a biologically significant target of 13b, positioning it as an advanced lead compound for further development of clinically relevant TACC3 inhibitors in cancers with elevated TACC3 expression.
Collapse
Affiliation(s)
- Deniz Lengerli
- Gazi University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 06560 Ankara, Turkey
| | - Özge Akbulut Çalışkan
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kübra Çalışkan
- Gazi University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 06560 Ankara, Turkey
| | - Özge Saatci
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Chaemin Lim
- A2A Pharmaceuticals, Inc., 1185 Avenue of the Americas, New York, NY 10036, USA
| | - Sridhar Vempati
- A2A Pharmaceuticals, Inc., 1185 Avenue of the Americas, New York, NY 10036, USA
| | - Burcu Çalışkan
- Gazi University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 06560 Ankara, Turkey
| | - Özgür Şahin
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Erden Banoglu
- Gazi University, Faculty of Pharmacy, Department of Pharmaceutical Chemistry, 06560 Ankara, Turkey.
| |
Collapse
|
3
|
Cammarata GM, Erdogan B, Sabo J, Kayaer Y, Dujava Zdimalova M, Engström F, Gupta U, Senel J, O'Brien T, Sibanda C, Thawani A, Folker ES, Braun M, Lansky Z, Lowery LA. The TOG5 domain of CKAP5 is required to interact with F-actin and promote microtubule advancement in neurons. Mol Biol Cell 2024; 35:br24. [PMID: 39504455 PMCID: PMC11656482 DOI: 10.1091/mbc.e24-05-0202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Microtubule (MT) and F-actin cytoskeletal cross-talk and organization are important aspects of axon guidance mechanisms, but how associated proteins facilitate this function remains largely unknown. While the MT-associated protein, CKAP5 (XMAP215/ch-TOG), has been best characterized as a MT polymerase, we have recently highlighted a novel role for CKAP5 in facilitating interactions between MT and F-actin in vitro and in embryonic Xenopus laevis neuronal growth cones. However, the mechanism by which it does so is unclear. Here, using in vitro reconstitution assays coupled with total internal reflection fluorescence microscopy, we report that the TOG5 domain of CKAP5 is necessary for its ability to bind to and bundle actin filaments, as well as to cross-link MTs and F-actin in vitro. Additionally, we show that this novel MT/F-actin cross-linking function of CKAP5 is possible even in MT polymerase-incompetent mutants of CKAP5 in vivo. Indeed, CKAP5 requires both MT and F-actin binding, but not MT polymerization, to promote MT-F-actin alignment in growth cones and axon outgrowth. Taken together, our findings provide mechanistic insights into how MT populations penetrate the growth cone periphery through CKAP5-facilitated interaction with F-actin during axon outgrowth and guidance.
Collapse
Affiliation(s)
| | - Burcu Erdogan
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Jan Sabo
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic 25240
| | - Yusuf Kayaer
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Michaela Dujava Zdimalova
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic 25240
- Department of Cell Biology, Faculty of Science, Charles University, Prague 12800, Czech Republic
| | - Filip Engström
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Urvika Gupta
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Jasming Senel
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Tara O'Brien
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Chiedza Sibanda
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| | - Akanksha Thawani
- Department of Molecular and Cell Biology, UC Berkeley, Berkeley CA 94720
| | - Eric S Folker
- Department of Biology, Boston College, Chestnut Hill, MA 02467
| | - Marcus Braun
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic 25240
| | - Zdenek Lansky
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, Vestec, Czech Republic 25240
| | - Laura A Lowery
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine and Boston Medical Center, Boston, MA 02118
| |
Collapse
|
4
|
Saatci O, Sahin O. TACC3: a multi-functional protein promoting cancer cell survival and aggressiveness. Cell Cycle 2023; 22:2637-2655. [PMID: 38197196 PMCID: PMC10936615 DOI: 10.1080/15384101.2024.2302243] [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: 11/01/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
TACC3 is the most oncogenic member of the transforming acidic coiled-coil domain-containing protein (TACC) family. It is one of the major recruitment factors of distinct multi-protein complexes. TACC3 is localized to spindles, centrosomes, and nucleus, and regulates key oncogenic processes, including cell proliferation, migration, invasion, and stemness. Recently, TACC3 inhibition has been identified as a vulnerability in highly aggressive cancers, such as cancers with centrosome amplification (CA). TACC3 has spatiotemporal functions throughout the cell cycle; therefore, targeting TACC3 causes cell death in mitosis and interphase in cancer cells with CA. In the clinics, TACC3 is highly expressed and associated with worse survival in multiple cancers. Furthermore, TACC3 is a part of one of the most common fusions of FGFR, FGFR3-TACC3 and is important for the oncogenicity of the fusion. A detailed understanding of the regulation of TACC3 expression, its key partners, and molecular functions in cancer cells is vital for uncovering the most vulnerable tumors and maximizing the therapeutic potential of targeting this highly oncogenic protein. In this review, we summarize the established and emerging interactors and spatiotemporal functions of TACC3 in cancer cells, discuss the potential of TACC3 as a biomarker in cancer, and therapeutic potential of its inhibition.
Collapse
Affiliation(s)
- Ozge Saatci
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
5
|
Hsueh HW, Kao HJ, Chao CC, Hsueh SJ, Huang YN, Lin WJ, Su JP, Shy HT, Yeh TY, Lin CC, Kwok PY, Lee NC, Hsieh ST. Identification of an 85-kb Heterozygous 4p Microdeletion With Full Genome Analysis in Autosomal Dominant Charcot-Marie-Tooth Disease. Neurol Genet 2023; 9:e200078. [PMID: 37346931 PMCID: PMC10281236 DOI: 10.1212/nxg.0000000000200078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/06/2023] [Indexed: 06/23/2023]
Abstract
Background and Objectives Charcot-Marie-Tooth disease (CMT) is a syndrome of a hereditary neurodegenerative condition affecting the peripheral nervous system and is a single gene disorder. Deep phenotyping coupled with advanced genetic techniques is critical in discovering new genetic defects of rare genetic disorders such as CMT. Methods We applied multidisciplinary investigations to examine the neurophysiology and nerve pathology in a family that fulfilled the diagnosis of CMT2. When phenotype-guided first-tier genetic tests and whole-exome sequencing did not yield a molecular diagnosis, we conducted full genome analysis by examining phased whole-genome sequencing and whole-genome optical mapping data to search for the causal variation. We then performed a systematic review to compare the reported patients with interstitial microdeletion in the short arm of chromosome 4. Results In this family with CMT2, we reported the discovery of a heterozygous 85-kb microdeletion in the short arm of chromosome 4 (4p16.3)[NC_000004.12:g.1733926_1819031del] spanning 3 genes [TACC3 (intron 6-exon 16), FGFR3 (total deletion), and LETM1 (intron 10-exon14)] that cosegregated with disease phenotypes in family members. The clinical features of peripheral nerve degeneration in our family are distinct from the well-known 4p microdeletion syndrome of Wolf-Hirschhorn syndrome, in which brain involvement is the major phenotype. Discussion In summary, we used the full genome analysis approach to discover a new microdeletion in a family with CMT2. The deleted segment contains 3 genes (TACC3, FGFR3, and LETM1) that likely play a role in the pathogenesis of nerve degeneration.
Collapse
Affiliation(s)
- Hsueh Wen Hsueh
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Hsiao-Jung Kao
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Chi-Chao Chao
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Sung-Ju Hsueh
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Yu-Ning Huang
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Wan-Jia Lin
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Jen-Ping Su
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Horng-Tzer Shy
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Ti-Yen Yeh
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Cheng-Chen Lin
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Pui-Yan Kwok
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Ni-Chung Lee
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| | - Sung-Tsang Hsieh
- From the Department of Neurology (H.W.H., C.-C.C., Y.-N.H., S.-T.H.), Department of Anatomy and Cell Biology (H.W.H., H.-T.S., T.-Y.Y., C.-C.L., S.-T.H.), National Taiwan University College of Medicine; Institute of Biomedical Sciences (H.-J.K., W.-J.L., J.-P.S., P.-Y.K.), Academia Sinica, Taipei; Department of Neurology (S.-J.H.), National Taiwan University Hospital Yunlin Branch; Institute for Human Genetics (P.-Y.K.), Cardiovascular Research Institute, and Department of Dermatology, University of California, San Francisco; and Department of Medical Genetics (N.-C.L.), National Taiwan University Hospital, Taipei
| |
Collapse
|
6
|
Atkins M, Nicol X, Fassier C. Microtubule remodelling as a driving force of axon guidance and pruning. Semin Cell Dev Biol 2023; 140:35-53. [PMID: 35710759 DOI: 10.1016/j.semcdb.2022.05.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/26/2022] [Accepted: 05/31/2022] [Indexed: 01/28/2023]
Abstract
The establishment of neuronal connectivity relies on the microtubule (MT) cytoskeleton, which provides mechanical support, roads for axonal transport and mediates signalling events. Fine-tuned spatiotemporal regulation of MT functions by tubulin post-translational modifications and MT-associated proteins is critical for the coarse wiring and subsequent refinement of neuronal connectivity. The defective regulation of these processes causes a wide range of neurodevelopmental disorders associated with connectivity defects. This review focuses on recent studies unravelling how MT composition, post-translational modifications and associated proteins influence MT functions in axon guidance and/or pruning to build functional neuronal circuits. We here summarise experimental evidence supporting the key role of this network as a driving force for growth cone steering and branch-specific axon elimination. We further provide a global overview of the MT-interactors that tune developing axon behaviours, with a special emphasis on their emerging versatility in the regulation of MT dynamics/structure. Recent studies establishing the key and highly selective role of the tubulin code in the regulation of MT functions in axon pathfinding are also reported. Finally, our review highlights the emerging molecular links between these MT regulation processes and guidance signals that wire the nervous system.
Collapse
Affiliation(s)
- Melody Atkins
- INSERM, UMR-S 1270, Institut du Fer à Moulin, Sorbonne Université, F-75005 Paris, France
| | - Xavier Nicol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Coralie Fassier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France.
| |
Collapse
|
7
|
Deng L, Zhao L, Jin J, Qiao B, Zhang X, Chang L, Zheng L, Dankar S, Ping J. Transforming acidic coiled-coil containing protein 3 suppresses influenza A virus replication by impeding viral endosomal trafficking and nuclear import. Vet Microbiol 2023; 282:109769. [PMID: 37148621 DOI: 10.1016/j.vetmic.2023.109769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/10/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023]
Abstract
Transforming acidic coiled-coil containing protein 3 (TACC3) is a motor spindle protein that plays an essential role in stabilization of the mitotic spindle. In this study, we show that the overexpression of TACC3 reduces the viral titers of multiple influenza A viruses (IAVs). In contrast, the downregulation of TACC3 increases IAVs propagation. Next, we map the target steps of TACC3 requirement to the early stages of viral replication. By confocal microscopy and nuclear plasma separation experiment, we reveal that overexpression of TACC3 results in a substantial decrease of IAV NP accumulation in the nuclei of infected cells. We further show that viral attachment and internalization are not affected by TACC3 overexpression and detect that the early and late endosomal trafficking of IAV in TACC3 overexpression cells is slower than negative control cells. These results suggest that TACC3 exerts an impaired effect on the endosomal trafficking and nuclear import of vRNP, thereby negatively regulating IAV replication. Moreover, the infection of different IAV subtypes decreases the expression level of TACC3 in turn. Consequently, we speculate that IAV ensures the generation of offspring virions by antagonizing the expression of inhibitory factor TACC3. Collectively, our results establish TACC3 as an important inhibitory factor for replication of the IAV, suggesting that TACC3 could be a potential target for the development of future antiviral compounds.
Collapse
Affiliation(s)
- Lulu Deng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Lingcai Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Jiayu Jin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Bingchen Qiao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaoting Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Lifeng Chang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Lucheng Zheng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Samar Dankar
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1V 8M5, Canada
| | - Jihui Ping
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Engineering Laboratory of Animal Immunity of Jiangsu Province, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
8
|
Gcap14 is a microtubule plus-end-tracking protein coordinating microtubule-actin crosstalk during neurodevelopment. Proc Natl Acad Sci U S A 2023; 120:e2214507120. [PMID: 36795749 PMCID: PMC9974511 DOI: 10.1073/pnas.2214507120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Regulation of microtubule dynamics is required to properly control various steps of neurodevelopment. In this study, we identified granule cell antiserum-positive 14 (Gcap14) as a microtubule plus-end-tracking protein and as a regulator of microtubule dynamics during neurodevelopment. Gcap14 knockout mice exhibited impaired cortical lamination. Gcap14 deficiency resulted in defective neuronal migration. Moreover, nuclear distribution element nudE-like 1 (Ndel1), an interacting partner of Gcap14, effectively corrected the downregulation of microtubule dynamics and the defects in neuronal migration caused by Gcap14 deficiency. Finally, we found that the Gcap14-Ndel1 complex participates in the functional link between microtubule and actin filament, thereby regulating their crosstalks in the growth cones of cortical neurons. Taken together, we propose that the Gcap14-Ndel1 complex is fundamental for cytoskeletal remodeling during neurodevelopmental processes such as neuronal processes elongation and neuronal migration.
Collapse
|
9
|
Vazquez-Pianzola P, Beuchle D, Saro G, Hernández G, Maldonado G, Brunßen D, Meister P, Suter B. Female meiosis II and pronuclear fusion require the microtubule transport factor Bicaudal D. Development 2022; 149:275749. [DOI: 10.1242/dev.199944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 05/25/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Bicaudal D (BicD) is a dynein adaptor that transports different cargoes along microtubules. Reducing the activity of BicD specifically in freshly laid Drosophila eggs by acute protein degradation revealed that BicD is needed to produce normal female meiosis II products, to prevent female meiotic products from re-entering the cell cycle, and for pronuclear fusion. Given that BicD is required to localize the spindle assembly checkpoint (SAC) components Mad2 and BubR1 to the female meiotic products, it appears that BicD functions to localize these components to control metaphase arrest of polar bodies. BicD interacts with Clathrin heavy chain (Chc), and both proteins localize to centrosomes, mitotic spindles and the tandem spindles during female meiosis II. Furthermore, BicD is required to localize clathrin and the microtubule-stabilizing factors transforming acidic coiled-coil protein (D-TACC/Tacc) and Mini spindles (Msps) correctly to the meiosis II spindles, suggesting that failure to localize these proteins may perturb SAC function. Furthermore, immediately after the establishment of the female pronucleus, D-TACC and Caenorhabditis elegans BicD, tacc and Chc are also needed for pronuclear fusion, suggesting that the underlying mechanism might be more widely used across species.
Collapse
Affiliation(s)
| | - Dirk Beuchle
- Institute of Cell Biology, University of Bern 1 , 3012 Berne , Switzerland
| | - Gabriella Saro
- Institute of Cell Biology, University of Bern 1 , 3012 Berne , Switzerland
| | - Greco Hernández
- Instituto Nacional de Cancerología (INCan) 2 Laboratory of Translation and Cancer, Unit of Biomedical Research on Cancer , , 14080-Tlalpan, Mexico City , Mexico
| | - Giovanna Maldonado
- Instituto Nacional de Cancerología (INCan) 2 Laboratory of Translation and Cancer, Unit of Biomedical Research on Cancer , , 14080-Tlalpan, Mexico City , Mexico
| | - Dominique Brunßen
- Institute of Cell Biology, University of Bern 1 , 3012 Berne , Switzerland
| | - Peter Meister
- Institute of Cell Biology, University of Bern 1 , 3012 Berne , Switzerland
| | - Beat Suter
- Institute of Cell Biology, University of Bern 1 , 3012 Berne , Switzerland
| |
Collapse
|
10
|
Meka DP, Kobler O, Hong S, Friedrich CM, Wuesthoff S, Henis M, Schwanke B, Krisp C, Schmuelling N, Rueter R, Ruecker T, Betleja E, Cheng T, Mahjoub MR, Soba P, Schlüter H, Fornasiero EF, Calderon de Anda F. Centrosome-dependent microtubule modifications set the conditions for axon formation. Cell Rep 2022; 39:110686. [PMID: 35443171 PMCID: PMC10150443 DOI: 10.1016/j.celrep.2022.110686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 12/27/2021] [Accepted: 03/24/2022] [Indexed: 11/29/2022] Open
Abstract
Microtubule (MT) modifications are critical during axon development, with stable MTs populating the axon. How these modifications are spatially coordinated is unclear. Here, via high-resolution microscopy, we show that early developing neurons have fewer somatic acetylated MTs restricted near the centrosome. At later stages, however, acetylated MTs spread out in soma and concentrate in growing axon. Live imaging in early plated neurons of the MT plus-end protein, EB3, show increased displacement and growth rate near the MTOC, suggesting local differences that might support axon selection. Moreover, F-actin disruption in early developing neurons, which show fewer somatic acetylated MTs, does not induce multiple axons, unlike later stages. Overexpression of centrosomal protein 120 (Cep120), which promotes MT acetylation/stabilization, induces multiple axons, while its knockdown downregulates proteins modulating MT dynamics and stability, hampering axon formation. Collectively, we show how centrosome-dependent MT modifications contribute to axon formation.
Collapse
Affiliation(s)
- Durga Praveen Meka
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Oliver Kobler
- Combinatorial Neuroimaging Core Facility, Leibniz Institute for Neurobiology, 39118 Magdeburg, Germany
| | - Shuai Hong
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Carina Meta Friedrich
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Souhaila Wuesthoff
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Melad Henis
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; Department of Anatomy and Histology, Faculty of Veterinary Medicine, New Valley University, 72511 El-Kharga, Egypt
| | - Birgit Schwanke
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Krisp
- Institute for Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics Group, Campus Forschung, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Nessa Schmuelling
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - René Rueter
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Tabitha Ruecker
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Ewelina Betleja
- Department of Medicine (Nephrology Division), Washington University, St. Louis, MO 63110, USA
| | - Tao Cheng
- Department of Medicine (Nephrology Division), Washington University, St. Louis, MO 63110, USA
| | - Moe R Mahjoub
- Department of Medicine (Nephrology Division), Washington University, St. Louis, MO 63110, USA
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, 53115 Bonn, Germany; Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Hartmut Schlüter
- Institute for Clinical Chemistry and Laboratory Medicine, Mass Spectrometric Proteomics Group, Campus Forschung, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Eugenio F Fornasiero
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Froylan Calderon de Anda
- Institute of Developmental Neurophysiology, Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| |
Collapse
|
11
|
Sánchez-Huertas C, Herrera E. With the Permission of Microtubules: An Updated Overview on Microtubule Function During Axon Pathfinding. Front Mol Neurosci 2021; 14:759404. [PMID: 34924953 PMCID: PMC8675249 DOI: 10.3389/fnmol.2021.759404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 11/01/2021] [Indexed: 01/27/2023] Open
Abstract
During the establishment of neural circuitry axons often need to cover long distances to reach remote targets. The stereotyped navigation of these axons defines the connectivity between brain regions and cellular subtypes. This chemotrophic guidance process mostly relies on the spatio-temporal expression patterns of extracellular proteins and the selective expression of their receptors in projection neurons. Axon guidance is stimulated by guidance proteins and implemented by neuronal traction forces at the growth cones, which engage local cytoskeleton regulators and cell adhesion proteins. Different layers of guidance signaling regulation, such as the cleavage and processing of receptors, the expression of co-receptors and a wide variety of intracellular cascades downstream of receptors activation, have been progressively unveiled. Also, in the last decades, the regulation of microtubule (MT) assembly, stability and interactions with the submembranous actin network in the growth cone have emerged as crucial effector mechanisms in axon pathfinding. In this review, we will delve into the intracellular signaling cascades downstream of guidance receptors that converge on the MT cytoskeleton of the growing axon. In particular, we will focus on the microtubule-associated proteins (MAPs) network responsible of MT dynamics in the axon and growth cone. Complementarily, we will discuss new evidences that connect defects in MT scaffold proteins, MAPs or MT-based motors and axon misrouting during brain development.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández (CSIC-UMH), Alicante, Spain
| | | |
Collapse
|
12
|
Furey C, Astar H, Walsh D. Human Cytomegalovirus Exploits TACC3 To Control Microtubule Dynamics and Late Stages of Infection. J Virol 2021; 95:e0082121. [PMID: 34191581 PMCID: PMC8387038 DOI: 10.1128/jvi.00821-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 06/25/2021] [Indexed: 01/23/2023] Open
Abstract
While it is well established that microtubules (MTs) facilitate various stages of virus replication, how viruses actively control MT dynamics and functions remains less well understood. Recent work has begun to reveal how several viruses exploit End-Binding (EB) proteins and their associated microtubule plus-end tracking proteins (+TIPs), in particular to enable loading of viral particles onto MTs for retrograde transport during early stages of infection. Distinct from other viruses studied to date, at mid- to late stages of its unusually protracted replication cycle, human cytomegalovirus (HCMV) increases the expression of all three EB family members. This occurs coincident with the formation of a unique structure, termed the assembly compartment (AC), which serves as a Golgi-derived MT organizing center. Together, the AC and distinct EB proteins enable HCMV to increase the formation of dynamic and acetylated microtubule subsets to regulate distinct aspects of the viral replication cycle. Here, we reveal that HCMV also exploits EB-independent +TIP pathways by specifically increasing the expression of transforming acidic coiled coil protein 3 (TACC3) to recruit the MT polymerase, chTOG, from initial sites of MT nucleation in the AC out into the cytosol, thereby increasing dynamic MT growth. Preventing TACC3 increases or depleting chTOG impaired MT polymerization, resulting in defects in early versus late endosome organization in and around the AC as well as defects in viral trafficking and spread. Our findings provide the first example of a virus that actively exploits EB-independent +TIP pathways to regulate MT dynamics and control late stages of virus replication. IMPORTANCE Diverse viruses rely on host cell microtubule networks to transport viral particles within the dense cytoplasmic environment and to control the broader architecture of the cell to facilitate their replication. However, precisely how viruses regulate the dynamic behavior and function of microtubule filaments remains poorly defined. We recently showed that the assembly compartment (AC) formed by human cytomegalovirus (HCMV) acts as a Golgi-derived microtubule organizing center. Here, we show that at mid- to late stages of infection, HCMV increases the expression of transforming acidic coiled coil protein 3 (TACC3) to control the localization of the microtubule polymerase, chTOG. This, in turn, enables HCMV to generate dynamic microtubule subsets that organize endocytic vesicles in and around the AC and facilitate the transport of new viral particles released into the cytosol. Our findings reveal the first instance of viral targeting of TACC3 to control microtubule dynamics and virus spread.
Collapse
Affiliation(s)
- Colleen Furey
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Helen Astar
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Derek Walsh
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
13
|
EFA6 in Axon Regeneration, as a Microtubule Regulator and as a Guanine Nucleotide Exchange Factor. Cells 2021; 10:cells10061325. [PMID: 34073530 PMCID: PMC8226579 DOI: 10.3390/cells10061325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 11/17/2022] Open
Abstract
Axon regeneration after injury is a conserved biological process that involves a large number of molecular pathways, including rapid calcium influx at injury sites, retrograde injury signaling, epigenetic transition, transcriptional reprogramming, polarized transport, and cytoskeleton reorganization. Despite the numerous efforts devoted to understanding the underlying cellular and molecular mechanisms of axon regeneration, the search continues for effective target molecules for improving axon regeneration. Although there have been significant historical efforts towards characterizing pro-regenerative factors involved in axon regeneration, the pursuit of intrinsic inhibitors is relatively recent. EFA6 (exchange factor for ARF6) has been demonstrated to inhibit axon regeneration in different organisms. EFA6 inhibition could be a promising therapeutic strategy to promote axon regeneration and functional recovery after axon injury. This review summarizes the inhibitory role on axon regeneration through regulating microtubule dynamics and through affecting ARF6 (ADP-ribosylation factor 6) GTPase-mediated integrin transport.
Collapse
|
14
|
Ryan EL, Shelford J, Massam-Wu T, Bayliss R, Royle SJ. Defining endogenous TACC3-chTOG-clathrin-GTSE1 interactions at the mitotic spindle using induced relocalization. J Cell Sci 2021; 134:jcs255794. [PMID: 33380489 PMCID: PMC7875487 DOI: 10.1242/jcs.255794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
A multiprotein complex containing TACC3, clathrin and other proteins has been implicated in mitotic spindle stability. To disrupt this complex in an anti-cancer context, we need to understand its composition and how it interacts with microtubules. Induced relocalization of proteins in cells is a powerful way to analyze protein-protein interactions and, additionally, monitor where and when these interactions occur. We used CRISPR/Cas9 gene editing to add tandem FKBP-GFP tags to each complex member. The relocalization of endogenous tagged protein from the mitotic spindle to mitochondria and assessment of the effect on other proteins allowed us to establish that TACC3 and clathrin are core complex members and that chTOG (also known as CKAP5) and GTSE1 are ancillary to the complex, binding respectively to TACC3 and clathrin, but not each other. We also show that PIK3C2A, a clathrin-binding protein that was proposed to stabilize the TACC3-chTOG-clathrin-GTSE1 complex during mitosis, is not a member of the complex. This work establishes that targeting the TACC3-clathrin interface or their microtubule-binding sites are the two strategies most likely to disrupt spindle stability mediated by this multiprotein complex.
Collapse
Affiliation(s)
- Ellis L Ryan
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - James Shelford
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Teresa Massam-Wu
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Gibbet Hill Road, Coventry CV4 7AL, UK
| | - Richard Bayliss
- School of Molecular and Cellular Biology, Astbury Centre for Structural Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Stephen J Royle
- Centre for Mechanochemical Cell Biology, Warwick Medical School, Gibbet Hill Road, Coventry CV4 7AL, UK
| |
Collapse
|
15
|
Furey C, Jovasevic V, Walsh D. TACC3 Regulates Microtubule Plus-End Dynamics and Cargo Transport in Interphase Cells. Cell Rep 2021; 30:269-283.e6. [PMID: 31914393 PMCID: PMC6980831 DOI: 10.1016/j.celrep.2019.12.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/13/2019] [Accepted: 12/06/2019] [Indexed: 12/12/2022] Open
Abstract
End-binding proteins (EBs) are widely viewed as master regulators of microtubule dynamics and function. Here, we show that while EB1 mediates the dynamic microtubule capture of herpes simplex virus type 1 (HSV-1) in fibroblasts, in neuronal cells, infection occurs independently of EBs through stable microtubules. Prompted by this, we find that transforming acid coiled-coil protein 3 (TACC3), widely studied in mitotic spindle formation, regulates the cytoplasmic localization of the microtubule polymerizing factor chTOG and influences microtubule plus-end dynamics during interphase to control infection in distinct cell types. Furthermore, perturbing TACC3 function in neuronal cells resulted in the formation of disorganized stable, detyrosinated microtubule networks and changes in cellular morphology, as well as impaired trafficking of both HSV-1 and transferrin. These trafficking defects in TACC3-depleted cells were reversed by the depletion of kinesin-1 heavy chains. As such, TACC3 is a critical regulator of interphase microtubule dynamics and stability that influences kinesin-1-based cargo trafficking. While EB proteins are widely studied as master regulators of microtubule plus-end dynamics, Furey et al. report EB-independent regulation of microtubule arrays and cargo trafficking by the transforming acid coiled-coil-containing protein, TACC3. By controlling the formation of detyrosinated stable microtubule networks, TACC3 influences kinesin-1-based sorting of both host and pathogenic cargoes.
Collapse
Affiliation(s)
- Colleen Furey
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Vladimir Jovasevic
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Derek Walsh
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
16
|
Rondelet A, Lin YC, Singh D, Porfetye AT, Thakur HC, Hecker A, Brinkert P, Schmidt N, Bendre S, Müller F, Mazul L, Widlund PO, Bange T, Hiller M, Vetter IR, Bird AW. Clathrin's adaptor interaction sites are repurposed to stabilize microtubules during mitosis. J Cell Biol 2020; 219:133599. [PMID: 31932847 PMCID: PMC7041688 DOI: 10.1083/jcb.201907083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/31/2019] [Accepted: 11/24/2019] [Indexed: 11/22/2022] Open
Abstract
Clathrin ensures mitotic spindle stability and efficient chromosome alignment, independently of its vesicle trafficking function. Although clathrin localizes to the mitotic spindle and kinetochore fiber microtubule bundles, the mechanisms by which clathrin stabilizes microtubules are unclear. We show that clathrin adaptor interaction sites on clathrin heavy chain (CHC) are repurposed during mitosis to directly recruit the microtubule-stabilizing protein GTSE1 to the spindle. Structural analyses reveal that these sites interact directly with clathrin-box motifs on GTSE1. Disruption of this interaction releases GTSE1 from spindles, causing defects in chromosome alignment. Surprisingly, this disruption destabilizes astral microtubules, but not kinetochore-microtubule attachments, and chromosome alignment defects are due to a failure of chromosome congression independent of kinetochore-microtubule attachment stability. GTSE1 recruited to the spindle by clathrin stabilizes microtubules by inhibiting the microtubule depolymerase MCAK. This work uncovers a novel role of clathrin adaptor-type interactions to stabilize nonkinetochore fiber microtubules to support chromosome congression, defining for the first time a repurposing of this endocytic interaction mechanism during mitosis.
Collapse
Affiliation(s)
- Arnaud Rondelet
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Yu-Chih Lin
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Divya Singh
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | | | - Harish C Thakur
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Andreas Hecker
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Pia Brinkert
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Nadine Schmidt
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Shweta Bendre
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | | | - Lisa Mazul
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Per O Widlund
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tanja Bange
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Michael Hiller
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany.,Center for Systems Biology, Dresden, Germany
| | - Ingrid R Vetter
- Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | | |
Collapse
|
17
|
Erdogan B, St Clair RM, Cammarata GM, Zaccaro T, Ballif BA, Lowery LA. Investigating the impact of the phosphorylation status of tyrosine residues within the TACC domain of TACC3 on microtubule behavior during axon growth and guidance. Cytoskeleton (Hoboken) 2020; 77:277-291. [PMID: 32543081 DOI: 10.1002/cm.21622] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 11/10/2022]
Abstract
Axon guidance is a critical process in forming the connections between a neuron and its target. The growth cone steers the growing axon toward the appropriate direction by integrating extracellular guidance cues and initiating intracellular signal transduction pathways downstream of these cues. The growth cone generates these responses by remodeling its cytoskeletal components. Regulation of microtubule dynamics within the growth cone is important for making guidance decisions. TACC3, as a microtubule plus-end binding (EB) protein, modulates microtubule dynamics during axon outgrowth and guidance. We have previously shown that Xenopus laevis embryos depleted of TACC3 displayed spinal cord axon guidance defects, while TACC3-overexpressing spinal neurons showed increased resistance to Slit2-induced growth cone collapse. Tyrosine kinases play an important role in relaying guidance signals to downstream targets during pathfinding events via inducing tyrosine phosphorylation. Here, in order to investigate the mechanism behind TACC3-mediated axon guidance, we examined whether tyrosine residues that are present in TACC3 have any role in regulating TACC3's interaction with microtubules or during axon outgrowth and guidance behaviors. We find that the phosphorylatable tyrosines within the TACC domain are important for the microtubule plus-end tracking behavior of TACC3. Moreover, TACC domain phosphorylation impacts axon outgrowth dynamics such as growth length and growth persistency. Together, our results suggest that tyrosine phosphorylation of TACC3 affects TACC3's microtubule plus-end tracking behavior as well as its ability to mediate axon growth dynamics in cultured embryonic neural tube explants.
Collapse
Affiliation(s)
- Burcu Erdogan
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA.,Harvard Department of Stem Cell and Regenerative Biology, Cambridge, Massachusetts, USA
| | - Riley M St Clair
- Department of Biology, University of Vermont, Burlington, Vermont, USA
| | | | - Timothy Zaccaro
- Department of Biology, Boston College, Chestnut Hill, Massachusetts, USA
| | - Bryan A Ballif
- Department of Biology, University of Vermont, Burlington, Vermont, USA
| | - Laura Anne Lowery
- Department of Medicine, Boston University Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Tang Q, Rui M, Bu S, Wang Y, Chew LY, Yu F. A microtubule polymerase is required for microtubule orientation and dendrite pruning in Drosophila. EMBO J 2020; 39:e103549. [PMID: 32267553 DOI: 10.15252/embj.2019103549] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 01/12/2023] Open
Abstract
Drosophila class IV ddaC neurons selectively prune all larval dendrites to refine the nervous system during metamorphosis. During dendrite pruning, severing of proximal dendrites is preceded by local microtubule (MT) disassembly. Here, we identify an unexpected role of Mini spindles (Msps), a conserved MT polymerase, in governing dendrite pruning. Msps associates with another MT-associated protein TACC, and both stabilize each other in ddaC neurons. Moreover, Msps and TACC are required to orient minus-end-out MTs in dendrites. We further show that the functions of msps in dendritic MT orientation and dendrite pruning are antagonized by the kinesin-13 MT depolymerase Klp10A. Excessive MT depolymerization, which is induced by pharmacological treatment and katanin overexpression, also perturbs dendritic MT orientation and dendrite pruning, phenocopying msps mutants. Thus, we demonstrate that the MT polymerase Msps is required to form dendritic minus-end-out MTs and thereby promotes dendrite pruning in Drosophila sensory neurons.
Collapse
Affiliation(s)
- Quan Tang
- Temasek Life Sciences Laboratory, Singapore City, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore City, Singapore
| | - Menglong Rui
- Temasek Life Sciences Laboratory, Singapore City, Singapore
| | - Shufeng Bu
- Temasek Life Sciences Laboratory, Singapore City, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore City, Singapore
| | - Yan Wang
- Temasek Life Sciences Laboratory, Singapore City, Singapore
| | - Liang Yuh Chew
- Temasek Life Sciences Laboratory, Singapore City, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore City, Singapore
| | - Fengwei Yu
- Temasek Life Sciences Laboratory, Singapore City, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore City, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, Centre for Life Sciences, Singapore City, Singapore.,Neuroscience and Behavioral Disorder Program, Duke-NUS Graduate Medical School Singapore, Singapore City, Singapore
| |
Collapse
|
19
|
Akbulut O, Lengerli D, Saatci O, Duman E, Seker UOS, Isik A, Akyol A, Caliskan B, Banoglu E, Sahin O. A Highly Potent TACC3 Inhibitor as a Novel Anticancer Drug Candidate. Mol Cancer Ther 2020; 19:1243-1254. [PMID: 32217742 DOI: 10.1158/1535-7163.mct-19-0957] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 02/11/2020] [Accepted: 03/19/2020] [Indexed: 11/16/2022]
Abstract
TACC3, a transforming acidic coiled-coil (TACC) family member, is frequently upregulated in a broad spectrum of cancers, including breast cancer. It plays critical roles in protecting microtubule stability and centrosome integrity that is often dysregulated in cancers; therefore, making TACC3 a highly attractive therapeutic target. Here, we identified a new TACC3-targeting chemotype, BO-264, through the screening of in-house compound collection. Direct interaction between BO-264 and TACC3 was validated by using several biochemical methods, including drug affinity responsive target stability, cellular thermal shift assay, and isothermal titration calorimetry. BO-264 demonstrated superior antiproliferative activity to the two currently reported TACC3 inhibitors, especially in aggressive breast cancer subtypes, basal and HER2+, via spindle assembly checkpoint-dependent mitotic arrest, DNA damage, and apoptosis, while the cytotoxicity against normal breast cells was negligible. Furthermore, BO-264 significantly decreased centrosomal TACC3 during both mitosis and interphase. BO-264 displayed potent antiproliferative activity (∼90% have less than 1 μmol/L GI50 value) in the NCI-60 cell line panel compromising of nine different cancer types. Noteworthy, BO-264 significantly inhibited the growth of cells harboring FGFR3-TACC3 fusion, an oncogenic driver in diverse malignancies. Importantly, its oral administration significantly impaired tumor growth in immunocompromised and immunocompetent breast and colon cancer mouse models, and increased survival without any major toxicity. Finally, TACC3 expression has been identified as strong independent prognostic factor in breast cancer and strongly prognostic in several different cancers. Overall, we identified a novel and highly potent TACC3 inhibitor as a novel potential anticancer agent, inducing spindle abnormalities and mitotic cell death.
Collapse
Affiliation(s)
- Ozge Akbulut
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey
| | - Deniz Lengerli
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Ozge Saatci
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey.,Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, South Carolina
| | - Elif Duman
- UNAM-National Nanotechnology Research Center, Institute of Material Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Urartu O S Seker
- UNAM-National Nanotechnology Research Center, Institute of Material Science and Nanotechnology, Bilkent University, Ankara, Turkey
| | - Aynur Isik
- Hacettepe University Transgenic Animal Technologies Research and Application Center, Ankara, Turkey
| | - Aytekin Akyol
- Hacettepe University Transgenic Animal Technologies Research and Application Center, Ankara, Turkey.,Department of Pathology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Burcu Caliskan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Erden Banoglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Ozgur Sahin
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Turkey. .,Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
20
|
Chou VT, Johnson S, Long J, Vounatsos M, Van Vactor D. dTACC restricts bouton addition and regulates microtubule organization at the Drosophila neuromuscular junction. Cytoskeleton (Hoboken) 2020; 77:4-15. [PMID: 31702858 PMCID: PMC7027520 DOI: 10.1002/cm.21578] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/11/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022]
Abstract
Regulation of the synaptic cytoskeleton is essential to proper neuronal development and wiring. Perturbations in neuronal microtubules (MTs) are associated with numerous pathologies, yet it remains unclear how changes in MTs may be coupled to synapse morphogenesis. Studies have identified many MT regulators that promote synapse growth. However, less is known about the factors that restrict growth, despite the potential links of synaptic overgrowth to severe neurological conditions. Here, we report that dTACC, which is implicated in MT assembly and stability, prevents synapse overgrowth at the Drosophila neuromuscular junction by restricting addition of new boutons throughout larval development. dTACC localizes to the axonal MT lattice and is required to maintain tubulin levels and the integrity of higher-order MT structures in motor axon terminals. While previous reports have demonstrated the roles of MT-stabilizing proteins in promoting synapse growth, our findings suggest that in certain contexts, MT stabilization may correlate with restricted growth.
Collapse
Affiliation(s)
- Vivian T. Chou
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| | - Seth Johnson
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| | - Jennifer Long
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| | - Maxime Vounatsos
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| | - David Van Vactor
- Department of Cell Biology and Program in NeuroscienceBlavatnik Institute, Harvard Medical SchoolBostonMassachusetts
| |
Collapse
|
21
|
Lasser M, Pratt B, Monahan C, Kim SW, Lowery LA. The Many Faces of Xenopus: Xenopus laevis as a Model System to Study Wolf-Hirschhorn Syndrome. Front Physiol 2019; 10:817. [PMID: 31297068 PMCID: PMC6607408 DOI: 10.3389/fphys.2019.00817] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 06/11/2019] [Indexed: 01/09/2023] Open
Abstract
Wolf–Hirschhorn syndrome (WHS) is a rare developmental disorder characterized by intellectual disability and various physical malformations including craniofacial, skeletal, and cardiac defects. These phenotypes, as they involve structures that are derived from the cranial neural crest, suggest that WHS may be associated with abnormalities in neural crest cell (NCC) migration. This syndrome is linked with assorted mutations on the short arm of chromosome 4, most notably the microdeletion of a critical genomic region containing several candidate genes. However, the function of these genes during embryonic development, as well as the cellular and molecular mechanisms underlying the disorder, are still unknown. The model organism Xenopus laevis offers a number of advantages for studying WHS. With the Xenopus genome sequenced, genetic manipulation strategies can be readily designed in order to alter the dosage of the WHS candidate genes. Moreover, a variety of assays are available for use in Xenopus to examine how manipulation of WHS genes leads to changes in the development of tissue and organ systems affected in WHS. In this review article, we highlight the benefits of using X. laevis as a model system for studying human genetic disorders of development, with a focus on WHS.
Collapse
Affiliation(s)
- Micaela Lasser
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Benjamin Pratt
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Connor Monahan
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Seung Woo Kim
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Laura Anne Lowery
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| |
Collapse
|
22
|
Slater PG, Cammarata GM, Samuelson AG, Magee A, Hu Y, Lowery LA. XMAP215 promotes microtubule-F-actin interactions to regulate growth cone microtubules during axon guidance in Xenopus laevis. J Cell Sci 2019; 132:jcs.224311. [PMID: 30890650 DOI: 10.1242/jcs.224311] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 03/08/2019] [Indexed: 12/24/2022] Open
Abstract
It has long been established that neuronal growth cone navigation depends on changes in microtubule (MT) and F-actin architecture downstream of guidance cues. However, the mechanisms by which MTs and F-actin are dually coordinated remain a fundamentally unresolved question. Here, we report that the well-characterized MT polymerase, XMAP215 (also known as CKAP5), plays an important role in mediating MT-F-actin interaction within the growth cone. We demonstrate that XMAP215 regulates MT-F-actin alignment through its N-terminal TOG 1-5 domains. Additionally, we show that XMAP215 directly binds to F-actin in vitro and co-localizes with F-actin in the growth cone periphery. We also find that XMAP215 is required for regulation of growth cone morphology and response to the guidance cue, Ephrin A5. Our findings provide the first strong evidence that XMAP215 coordinates MT and F-actin interaction in vivo We suggest a model in which XMAP215 regulates MT extension along F-actin bundles into the growth cone periphery and that these interactions may be important to control cytoskeletal dynamics downstream of guidance cues. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Paula G Slater
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | | | | | - Alexandra Magee
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | - Yuhan Hu
- Department of Cell Biology, Yale University, New Haven, CT 06520, USA
| | | |
Collapse
|
23
|
Mills A, Bearce E, Cella R, Kim SW, Selig M, Lee S, Lowery LA. Wolf-Hirschhorn Syndrome-Associated Genes Are Enriched in Motile Neural Crest Cells and Affect Craniofacial Development in Xenopus laevis. Front Physiol 2019; 10:431. [PMID: 31031646 PMCID: PMC6474402 DOI: 10.3389/fphys.2019.00431] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/28/2019] [Indexed: 01/08/2023] Open
Abstract
Wolf-Hirschhorn Syndrome (WHS) is a human developmental disorder arising from a hemizygous perturbation, typically a microdeletion, on the short arm of chromosome four. In addition to pronounced intellectual disability, seizures, and delayed growth, WHS presents with a characteristic facial dysmorphism and varying prevalence of microcephaly, micrognathia, cartilage malformation in the ear and nose, and facial asymmetries. These affected craniofacial tissues all derive from a shared embryonic precursor, the cranial neural crest (CNC), inviting the hypothesis that one or more WHS-affected genes may be critical regulators of neural crest development or migration. To explore this, we characterized expression of multiple genes within or immediately proximal to defined WHS critical regions, across the span of craniofacial development in the vertebrate model system Xenopus laevis. This subset of genes, whsc1, whsc2, letm1, and tacc3, are diverse in their currently-elucidated cellular functions; yet we find that their expression demonstrates shared tissue-specific enrichment within the anterior neural tube, migratory neural crest, and later craniofacial structures. We examine the ramifications of this by characterizing craniofacial development and neural crest migration following individual gene depletion. We observe that several WHS-associated genes significantly impact facial patterning, cartilage formation, neural crest motility in vivo and in vitro, and can separately contribute to forebrain scaling. Thus, we have determined that numerous genes within and surrounding the defined WHS critical regions potently impact craniofacial patterning, suggesting their role in WHS presentation may stem from essential functions during neural crest-derived tissue formation.
Collapse
Affiliation(s)
- Alexandra Mills
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Elizabeth Bearce
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Rachael Cella
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Seung Woo Kim
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Megan Selig
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Sangmook Lee
- Biology Department, Boston College, Chestnut Hill, MA, United States
| | - Laura Anne Lowery
- Biology Department, Boston College, Chestnut Hill, MA, United States
| |
Collapse
|
24
|
Slater PG, Cammarata GM, Monahan C, Bowers JT, Yan O, Lee S, Lowery LA. Characterization of Xenopus laevis guanine deaminase reveals new insights for its expression and function in the embryonic kidney. Dev Dyn 2019; 248:296-305. [PMID: 30682232 DOI: 10.1002/dvdy.14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/18/2018] [Accepted: 01/21/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The mammalian guanine deaminase (GDA), called cypin, is important for proper neural development, by regulating dendritic arborization through modulation of microtubule (MT) dynamics. Additionally, cypin can promote MT assembly in vitro. However, it has never been tested whether cypin (or other GDA orthologs) binds to MTs or modulates MT dynamics. Here, we address these questions and characterize Xenopus laevis GDA (Gda) for the first time during embryonic development. RESULTS We find that exogenously expressed human cypin and Gda both display a cytosolic distribution in primary embryonic cells. Furthermore, while expression of human cypin can promote MT polymerization, Xenopus Gda has no effect. Additionally, we find that the tubulin-binding collapsin response mediator protein (CRMP) homology domain is only partially conserved between cypin and Gda. This likely explains the divergence in function, as we discovered that the cypin region containing the CRMP homology and PDZ-binding domain is necessary for regulating MT dynamics. Finally, we observed that gda is strongly expressed in the kidneys during late embryonic development, although it does not appear to be critical for kidney development. CONCLUSIONS Together, these results suggest that GDA has diverged in function between mammals and amphibians, and that mammalian GDA plays an indirect role in regulating MT dynamics. Developmental Dynamics 248:296-305, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Paula G Slater
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | | | - Connor Monahan
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | - Jackson T Bowers
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | - Oliver Yan
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | - Sangmook Lee
- Boston College, Department of Biology, Chestnut Hill, Massachusetts
| | | |
Collapse
|
25
|
IGARASHI M. Molecular basis of the functions of the mammalian neuronal growth cone revealed using new methods. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2019; 95:358-377. [PMID: 31406059 PMCID: PMC6766448 DOI: 10.2183/pjab.95.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 04/26/2019] [Indexed: 05/25/2023]
Abstract
The neuronal growth cone is a highly motile, specialized structure for extending neuronal processes. This structure is essential for nerve growth, axon pathfinding, and accurate synaptogenesis. Growth cones are important not only during development but also for plasticity-dependent synaptogenesis and neuronal circuit rearrangement following neural injury in the mature brain. However, the molecular details of mammalian growth cone function are poorly understood. This review examines molecular findings on the function of the growth cone as a result of the introduction of novel methods such superresolution microscopy and (phospho)proteomics. These results increase the scope of our understating of the molecular mechanisms of growth cone behavior in the mammalian brain.
Collapse
Affiliation(s)
- Michihiro IGARASHI
- Department of Neurochemistry and Molecular Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
26
|
Lasser M, Tiber J, Lowery LA. The Role of the Microtubule Cytoskeleton in Neurodevelopmental Disorders. Front Cell Neurosci 2018; 12:165. [PMID: 29962938 PMCID: PMC6010848 DOI: 10.3389/fncel.2018.00165] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/28/2018] [Indexed: 12/28/2022] Open
Abstract
Neurons depend on the highly dynamic microtubule (MT) cytoskeleton for many different processes during early embryonic development including cell division and migration, intracellular trafficking and signal transduction, as well as proper axon guidance and synapse formation. The coordination and support from MTs is crucial for newly formed neurons to migrate appropriately in order to establish neural connections. Once connections are made, MTs provide structural integrity and support to maintain neural connectivity throughout development. Abnormalities in neural migration and connectivity due to genetic mutations of MT-associated proteins can lead to detrimental developmental defects. Growing evidence suggests that these mutations are associated with many different neurodevelopmental disorders, including intellectual disabilities (ID) and autism spectrum disorders (ASD). In this review article, we highlight the crucial role of the MT cytoskeleton in the context of neurodevelopment and summarize genetic mutations of various MT related proteins that may underlie or contribute to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Micaela Lasser
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Jessica Tiber
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| | - Laura Anne Lowery
- Department of Biology, Boston College, Chestnut Hill, MA, United States
| |
Collapse
|
27
|
NEK7 regulates dendrite morphogenesis in neurons via Eg5-dependent microtubule stabilization. Nat Commun 2018; 9:2330. [PMID: 29899413 PMCID: PMC5997995 DOI: 10.1038/s41467-018-04706-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/15/2018] [Indexed: 01/22/2023] Open
Abstract
Organization of microtubules into ordered arrays is best understood in mitotic systems, but remains poorly characterized in postmitotic cells such as neurons. By analyzing the cycling cell microtubule cytoskeleton proteome through expression profiling and targeted RNAi screening for candidates with roles in neurons, we have identified the mitotic kinase NEK7. We show that NEK7 regulates dendrite morphogenesis in vitro and in vivo. NEK7 kinase activity is required for dendrite growth and branching, as well as spine formation and morphology. NEK7 regulates these processes in part through phosphorylation of the kinesin Eg5/KIF11, promoting its accumulation on microtubules in distal dendrites. Here, Eg5 limits retrograde microtubule polymerization, which is inhibitory to dendrite growth and branching. Eg5 exerts this effect through microtubule stabilization, independent of its motor activity. This work establishes NEK7 as a general regulator of the microtubule cytoskeleton, controlling essential processes in both mitotic cells and postmitotic neurons. NEK7 is a kinase known for its role in mitotic spindle assembly, driving centrosome separation in prophase through regulation of the kinesin Eg5. Here, the authors show that NEK7 and Eg5 also control dendrite morphogenesis in postmitotic neurons.
Collapse
|
28
|
Chen L. Microtubules and axon regeneration in C. elegans. Mol Cell Neurosci 2018; 91:160-166. [PMID: 29551667 DOI: 10.1016/j.mcn.2018.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 11/28/2022] Open
Abstract
Axon regeneration is a fundamental and conserved process that allows the nervous system to repair circuits after trauma. Due to its conserved genome, transparent body, and relatively simple neuroanatomy, C. elegans has become a powerful model organism for studying the cellular and molecular mechanisms underlying axon regeneration. Various studies from different model organisms have found microtubule dynamics to be pivotal to axon regrowth. In this review, we will discuss the latest findings on how microtubule dynamics are regulated during axon regeneration in C. elegans. Understanding the mechanisms of axon regeneration will aid in the development of more effective therapeutic strategies for treatments of diseases involving disconnection of axons, such as spinal cord injury and stroke.
Collapse
Affiliation(s)
- Lizhen Chen
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, Department of Molecular Medicine, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
29
|
Ding ZM, Huang CJ, Jiao XF, Wu D, Huo LJ. The role of TACC3 in mitotic spindle organization. Cytoskeleton (Hoboken) 2017; 74:369-378. [PMID: 28745816 DOI: 10.1002/cm.21388] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 07/04/2017] [Accepted: 07/21/2017] [Indexed: 12/31/2022]
Abstract
TACC3 regulates spindle organization during mitosis and also regulates centrosome-mediated microtubule nucleation by affecting γ-Tubulin ring complexes. In addition, it interacts with different proteins (such as ch-TOG, clathrin and Aurora-A) to function in mitotic spindle assembly and stability. By forming the TACC3/ch-TOG complex, TACC3 acts as a plus end-tracking protein to promote microtubule elongation. The TACC3/ch-TOG/clathrin complex is formed to stabilize kinetochore fibers by crosslinking adjacent microtubules. Furthermore, the phosphorylation of TACC3 by Aurora-A is important for the formation of TACC3/ch-TOG/clathrin and its recruitment to kinetochore fibers. Recently, the aberrant expression of TACC3 in a variety of human cancers has been linked with mitotic defects. Thus, in this review, we will discuss our current understanding of the biological roles of TACC3 in mitotic spindle organization.
Collapse
Affiliation(s)
- Zhi-Ming Ding
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| | - Chun-Jie Huang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| | - Xiao-Fei Jiao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| | - Di Wu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| | - Li-Jun Huo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Education Ministry of China, College of Animal Science and Technology, Huazhong, Agricultural University, Wuhan, 430070, China
| |
Collapse
|
30
|
Slater PG, Hayrapetian L, Lowery LA. Xenopus laevis as a model system to study cytoskeletal dynamics during axon pathfinding. Genesis 2017; 55. [PMID: 28095612 DOI: 10.1002/dvg.22994] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 11/02/2016] [Accepted: 11/04/2016] [Indexed: 01/17/2023]
Abstract
The model system, Xenopus laevis, has been used in innumerable research studies and has contributed to the understanding of multiple cytoskeletal components, including actin, microtubules, and neurofilaments, during axon pathfinding. Xenopus developmental stages have been widely characterized, and the Xenopus genome has been sequenced, allowing gene expression modifications through exogenous molecules. Xenopus cell cultures are ideal for long periods of live imaging because they are easily obtained and maintained, and they do not require special culture conditions. In addition, Xenopus have relatively large growth cones, compared to other vertebrates, thus providing a suitable system for imaging cytoskeletal components. Therefore, X. laevis is an ideal model organism in which to study cytoskeletal dynamics during axon pathfinding.
Collapse
Affiliation(s)
- Paula G Slater
- Department of Biology, Boston College, Chestnut Hill, Massachusetts
| | | | | |
Collapse
|
31
|
Erdogan B, Cammarata GM, Lee EJ, Pratt BC, Francl AF, Rutherford EL, Lowery LA. The microtubule plus-end-tracking protein TACC3 promotes persistent axon outgrowth and mediates responses to axon guidance signals during development. Neural Dev 2017; 12:3. [PMID: 28202041 PMCID: PMC5312526 DOI: 10.1186/s13064-017-0080-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 02/06/2017] [Indexed: 12/14/2022] Open
Abstract
Background Formation of precise neuronal connections requires proper axon guidance. Microtubules (MTs) of the growth cone provide a critical driving force during navigation of the growing ends of axons. Pioneer MTs and their plus-end tracking proteins (+TIPs) are thought to play integrative roles during this navigation. TACC3 is a + TIP that we have previously implicated in regulating MT dynamics within axons. However, the role of TACC3 in axon guidance has not been previously explored. Results Here, we show that TACC3 is required to promote persistent axon outgrowth and prevent spontaneous axon retractions in embryonic Xenopus laevis neurons. We also show that overexpressing TACC3 can counteract the depolymerizing effect of low doses of nocodazole, and that TACC3 interacts with MT polymerase XMAP215 to promote axon outgrowth. Moreover, we demonstrate that manipulation of TACC3 levels interferes with the growth cone response to the axon guidance cue Slit2 ex vivo, and that ablation of TACC3 causes pathfinding defects in axons of developing spinal neurons in vivo. Conclusion Together, our results suggest that by mediating MT dynamics, the + TIP TACC3 is involved in axon outgrowth and pathfinding decisions of neurons during embryonic development.
Collapse
Affiliation(s)
- Burcu Erdogan
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | | | - Eric J Lee
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Benjamin C Pratt
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Andrew F Francl
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Erin L Rutherford
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA
| | - Laura Anne Lowery
- Department of Biology, Boston College, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
32
|
Rutherford EL, Lowery LA. Exploring the developmental mechanisms underlying Wolf-Hirschhorn Syndrome: Evidence for defects in neural crest cell migration. Dev Biol 2016; 420:1-10. [PMID: 27777068 PMCID: PMC5193094 DOI: 10.1016/j.ydbio.2016.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/03/2016] [Accepted: 10/18/2016] [Indexed: 01/20/2023]
Abstract
Wolf-Hirschhorn Syndrome (WHS) is a neurodevelopmental disorder characterized by mental retardation, craniofacial malformation, and defects in skeletal and heart development. The syndrome is associated with irregularities on the short arm of chromosome 4, including deletions of varying sizes and microduplications. Many of these genotypic aberrations in humans have been correlated with the classic WHS phenotype, and animal models have provided a context for mapping these genetic irregularities to specific phenotypes; however, there remains a significant knowledge gap concerning the cell biological mechanisms underlying these phenotypes. This review summarizes literature that has made recent contributions to this topic, drawing from the vast body of knowledge detailing the genetic particularities of the disorder and the more limited pool of information on its cell biology. Finally, we propose a novel characterization for WHS as a pathophysiology owing in part to defects in neural crest cell motility and migration during development.
Collapse
Affiliation(s)
- Erin L Rutherford
- Boston College, Department of Biology, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, United States
| | - Laura Anne Lowery
- Boston College, Department of Biology, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, United States.
| |
Collapse
|
33
|
Voelzmann A, Hahn I, Pearce SP, Sánchez-Soriano N, Prokop A. A conceptual view at microtubule plus end dynamics in neuronal axons. Brain Res Bull 2016; 126:226-237. [PMID: 27530065 PMCID: PMC5090033 DOI: 10.1016/j.brainresbull.2016.08.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 12/02/2022]
Abstract
Axons are the cable-like protrusions of neurons which wire up the nervous system. Polar bundles of microtubules (MTs) constitute their structural backbones and are highways for life-sustaining transport between proximal cell bodies and distal synapses. Any morphogenetic changes of axons during development, plastic rearrangement, regeneration or degeneration depend on dynamic changes of these MT bundles. A key mechanism for implementing such changes is the coordinated polymerisation and depolymerisation at the plus ends of MTs within these bundles. To gain an understanding of how such regulation can be achieved at the cellular level, we provide here an integrated overview of the extensive knowledge we have about the molecular mechanisms regulating MT de/polymerisation. We first summarise insights gained from work in vitro, then describe the machinery which supplies the essential tubulin building blocks, the protein complexes associating with MT plus ends, and MT shaft-based mechanisms that influence plus end dynamics. We briefly summarise the contribution of MT plus end dynamics to important cellular functions in axons, and conclude by discussing the challenges and potential strategies of integrating the existing molecular knowledge into conceptual understanding at the level of axons.
Collapse
Affiliation(s)
- André Voelzmann
- The University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Ines Hahn
- The University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK
| | - Simon P Pearce
- The University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK; The University of Manchester, School of Mathematics, Alan Turing Building, Oxford Road, Manchester M13 9PL, UK
| | - Natalia Sánchez-Soriano
- University of Liverpool, Institute of Translational Medicine, Department of Cellular and Molecular Physiology, Crown Street, Liverpool, L69 3BX, UK
| | - Andreas Prokop
- The University of Manchester, Faculty of Biology, Medicine and Health, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
34
|
Murata S, Zhang C, Finch N, Zhang K, Campo L, Breuer EK. Predictors and Modulators of Synthetic Lethality: An Update on PARP Inhibitors and Personalized Medicine. BIOMED RESEARCH INTERNATIONAL 2016; 2016:2346585. [PMID: 27642590 PMCID: PMC5013223 DOI: 10.1155/2016/2346585] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 07/28/2016] [Indexed: 12/18/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP) inhibitors have proven to be successful agents in inducing synthetic lethality in several malignancies. Several PARP inhibitors have reached clinical trial testing for treatment in different cancers, and, recently, Olaparib (AZD2281) has gained both United States Food and Drug Administration (USFDA) and the European Commission (EC) approval for use in BRCA-mutated advanced ovarian cancer treatment. The need to identify biomarkers, their interactions in DNA damage repair pathways, and their potential utility in identifying patients who are candidates for PARP inhibitor treatment is well recognized. In this review, we detail many of the biomarkers that have been investigated for their ability to predict both PARP inhibitor sensitivity and resistance in preclinical studies as well as the results of several clinical trials that have tested the safety and efficacy of different PARP inhibitor agents in BRCA and non-BRCA-mutated cancers.
Collapse
Affiliation(s)
- Stephen Murata
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Catherine Zhang
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Nathan Finch
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Kevin Zhang
- Department of Otorhinolaryngology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Loredana Campo
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Eun-Kyoung Breuer
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
35
|
Rutherford EL, Carandang L, Ebbert PT, Mills AN, Bowers JT, Lowery LA. Xenopus TACC2 is a microtubule plus end-tracking protein that can promote microtubule polymerization during embryonic development. Mol Biol Cell 2016; 27:3013-3020. [PMID: 27559128 PMCID: PMC5063610 DOI: 10.1091/mbc.e16-03-0198] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/17/2016] [Indexed: 11/23/2022] Open
Abstract
Xenopus TACC2 is a microtubule plus end–tracking protein that localizes in front of EB1 and overlaps with TACC1 and TACC3 in cultured embryonic mesenchymal cells and neuronal growth cones. TACC2 OE can promote increased microtubule polymerization in mesenchymal cells but not growth cones, suggesting cell-type specificity to its function. Microtubule dynamics is regulated by plus end–tracking proteins (+TIPs), which localize to the plus ends of microtubules (MTs). We previously showed that TACC1 and TACC3, members of the transforming acidic coiled-coil protein family, can act as +TIPs to regulate MT dynamics in Xenopus laevis. Here we characterize TACC2 as a +TIP that localizes to MT plus ends in front of EB1 and overlapping with TACC1 and TACC3 in multiple embryonic cell types. We also show that TACC2 can promote MT polymerization in mesenchymal cells but not neuronal growth cones, thus displaying cell-type specificity. Structure–function analysis demonstrates that the C-terminal region of TACC2 is both necessary and sufficient to localize to MT plus ends and promote increased rates of MT polymerization, whereas the N-terminal region cannot bind to MT plus ends but can act in a dominant-negative capacity to reduce polymerization rates. Finally, we analyze mRNA expression patterns in Xenopus embryos for each TACC protein and observe neural enrichment of TACC3 expression compared with TACC1 and TACC2, which are also expressed in mesodermal tissues, including somites. Overall these data provide a novel assessment of all three TACC proteins as a family of +TIPs by highlighting the unique attributes of each, as well as their collective characteristics.
Collapse
|
36
|
Zhu X, Cao Q, Li X, Wang Z. Knockdown of TACC3 inhibits trophoblast cell migration and invasion through the PI3K/Akt signaling pathway. Mol Med Rep 2016; 14:3437-42. [PMID: 27572091 DOI: 10.3892/mmr.2016.5659] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 08/01/2016] [Indexed: 11/06/2022] Open
Abstract
The insufficient invasion of trophoblasts is known to be correlated with the development of preeclampsia. Transforming acidic coiled‑coil protein 3 (TACC3), a member of the TACC domain family, is important in the regulation of cell differentiation, migration and invasion. However, the role of TACC3 in trophoblast function during placental development remains to be fully elucidated. The present study aimed to determine the expression and function of TACC3 in human placenta and to examine the underlying mechanisms. TACC3 expression was analyzed in preeclamptic placentas using reverse transcription‑quantitative polymerase chain reaction and western blotting. Cell proliferation was determined by the MTT assay, and cell migration and invasion were measured using Transwell assays. The expression levels of TACC3, matrix metalloproteinase (MMP)‑2, MMP‑9, tissue inhibitor of metalloproteinase (TIMP)‑1, TIMP‑2, phosphoinositide 3‑kinase (PI3K), phosphorylated (p)‑PI3K, AKT and p‑AKT were detected by western blotting. The results showed that the expression of TACC3 was downregulated in preeclamptic placentas. The knockdown of TACC3 significantly inhibited HTR8/SVneo cell proliferation, migration and invasion, and inhibited the expression of matrix metalloproteinases. In addition, the knockdown of TACC3 significantly reduced the levels of p‑PI3K and Akt in the HTR8/SVneo cells. Taken together, the results of the present study demonstrated that the knockdown of TACC3 inhibited the migration and invasion of HTR8/SVneo cells through suppression of the PI3K/Akt signaling pathway. Therefore, TACC3 may serve as a novel potential target for treating preeclampsia.
Collapse
Affiliation(s)
- Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Qianqian Cao
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Xia Li
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhengping Wang
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
37
|
van de Willige D, Hoogenraad CC, Akhmanova A. Microtubule plus-end tracking proteins in neuronal development. Cell Mol Life Sci 2016; 73:2053-77. [PMID: 26969328 PMCID: PMC4834103 DOI: 10.1007/s00018-016-2168-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/04/2016] [Accepted: 02/22/2016] [Indexed: 11/28/2022]
Abstract
Regulation of the microtubule cytoskeleton is of pivotal importance for neuronal development and function. One such regulatory mechanism centers on microtubule plus-end tracking proteins (+TIPs): structurally and functionally diverse regulatory factors, which can form complex macromolecular assemblies at the growing microtubule plus-ends. +TIPs modulate important properties of microtubules including their dynamics and their ability to control cell polarity, membrane transport and signaling. Several neurodevelopmental and neurodegenerative diseases are associated with mutations in +TIPs or with misregulation of these proteins. In this review, we focus on the role and regulation of +TIPs in neuronal development and associated disorders.
Collapse
Affiliation(s)
- Dieudonnée van de Willige
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| |
Collapse
|
38
|
Abstract
The capacity of an axon to regenerate is regulated by its external environment and by cell-intrinsic factors. Studies in a variety of organisms suggest that alterations in axonal microtubule (MT) dynamics have potent effects on axon regeneration. We review recent findings on the regulation of MT dynamics during axon regeneration, focusing on the nematode Caenorhabditis elegans. In C. elegans the dual leucine zipper kinase (DLK) promotes axon regeneration, whereas the exchange factor for Arf6 (EFA-6) inhibits axon regeneration. Both DLK and EFA-6 respond to injury and control axon regeneration in part via MT dynamics. How the DLK and EFA-6 pathways are related is a topic of active investigation, as is the mechanism by which EFA-6 responds to axonal injury. We evaluate potential candidates, such as the MT affinity-regulating kinase PAR-1/MARK, in regulation of EFA-6 and axonal MT dynamics in regeneration.
Collapse
Affiliation(s)
- Ngang Heok Tang
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Andrew D Chisholm
- Section of Neurobiology, Division of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
39
|
Lucaj CM, Evans MF, Nwagbara BU, Ebbert PT, Baker CC, Volk JG, Francl AF, Ruvolo SP, Lowery LA. Xenopus TACC1 is a microtubule plus-end tracking protein that can regulate microtubule dynamics during embryonic development. Cytoskeleton (Hoboken) 2016; 72:225-34. [PMID: 26012630 PMCID: PMC4520305 DOI: 10.1002/cm.21224] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/29/2015] [Accepted: 05/13/2015] [Indexed: 12/30/2022]
Abstract
Microtubule plus‐end dynamics are regulated by a family of proteins called plus‐end tracking proteins (+TIPs). We recently demonstrated that the transforming acidic coiled‐coil (TACC) domain family member, TACC3, can function as a +TIP to regulate microtubule dynamics in Xenopus laevis embryonic cells. Although it has been previously reported that TACC3 is the only TACC family member that exists in Xenopus, our examination of its genome determined that Xenopus, like all other vertebrates, contains three TACC family members. Here, we investigate the localization and function of Xenopus TACC1, the founding member of the TACC family. We demonstrate that it can act as a +TIP to regulate microtubule dynamics, and that the conserved C‐terminal TACC domain is required for its localization to plus‐ends. We also show that, in Xenopus embryonic mesenchymal cells, TACC1 and TACC3 are each required for maintaining normal microtubule growth speed but exhibit some functional redundancy in the regulation of microtubule growth lifetime. Given the conservation of TACC1 in Xenopus and other vertebrates, we propose that Xenopus laevis is a useful system to investigate unexplored cell biological functions of TACC1 and other TACC family members in the regulation of microtubule dynamics. © 2015 The Authors. Cytoskeleton, Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Christopher M Lucaj
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| | - Matthew F Evans
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| | - Belinda U Nwagbara
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| | - Patrick T Ebbert
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| | - Charlie C Baker
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| | - Joseph G Volk
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| | - Andrew F Francl
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| | - Sean P Ruvolo
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| | - Laura Anne Lowery
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, Massachusetts
| |
Collapse
|
40
|
Cammarata GM, Bearce EA, Lowery LA. Cytoskeletal social networking in the growth cone: How +TIPs mediate microtubule-actin cross-linking to drive axon outgrowth and guidance. Cytoskeleton (Hoboken) 2016; 73:461-76. [PMID: 26783725 DOI: 10.1002/cm.21272] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 01/08/2016] [Accepted: 01/08/2016] [Indexed: 11/08/2022]
Abstract
The growth cone is a unique structure capable of guiding axons to their proper destinations. Within the growth cone, extracellular guidance cues are interpreted and then transduced into physical changes in the actin filament (F-actin) and microtubule cytoskeletons, providing direction and movement. While both cytoskeletal networks individually possess important growth cone-specific functions, recent data over the past several years point towards a more cooperative role between the two systems. Facilitating this interaction between F-actin and microtubules, microtubule plus-end tracking proteins (+TIPs) have been shown to link the two cytoskeletons together. Evidence suggests that many +TIPs can couple microtubules to F-actin dynamics, supporting both microtubule advance and retraction in the growth cone periphery. In addition, growing in vitro and in vivo data support a secondary role for +TIPs in which they may participate as F-actin nucleators, thus directly influencing F-actin dynamics and organization. This review focuses on how +TIPs may link F-actin and microtubules together in the growth cone, and how these interactions may influence axon guidance. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | | | - Laura Anne Lowery
- Department of Biology, Boston College, Chestnut Hill, Massachusetts.
| |
Collapse
|
41
|
Erdogan B, Ebbert PT, Lowery LA. Using Xenopus laevis retinal and spinal neurons to study mechanisms of axon guidance in vivo and in vitro. Semin Cell Dev Biol 2016; 51:64-72. [PMID: 26853934 DOI: 10.1016/j.semcdb.2016.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/01/2016] [Indexed: 11/26/2022]
Abstract
The intricate and precise establishment of neuronal connections in the developing nervous system relies on accurate navigation of growing axons. Since Ramón y Cajal's discovery of the growth cone, the phenomenon of axon guidance has been revealed as a coordinated operation of guidance molecules, receptors, secondary messengers, and responses driven by the dynamic cytoskeleton within the growth cone. With the advent of new and accelerating techniques, Xenopus laevis emerged as a robust model to investigate neuronal circuit formation during development. We present here the advantages of the Xenopus nervous system to our growing understanding of axon guidance.
Collapse
Affiliation(s)
- Burcu Erdogan
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA.
| | - Patrick T Ebbert
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA.
| | - Laura Anne Lowery
- Department of Biology, Boston College, 140 Commonwealth Avenue, Chestnut Hill, MA 02467, USA.
| |
Collapse
|
42
|
Cavazza T, Vernos I. The RanGTP Pathway: From Nucleo-Cytoplasmic Transport to Spindle Assembly and Beyond. Front Cell Dev Biol 2016; 3:82. [PMID: 26793706 PMCID: PMC4707252 DOI: 10.3389/fcell.2015.00082] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 12/07/2015] [Indexed: 01/03/2023] Open
Abstract
The small GTPase Ran regulates the interaction of transport receptors with a number of cellular cargo proteins. The high affinity binding of the GTP-bound form of Ran to import receptors promotes cargo release, whereas its binding to export receptors stabilizes their interaction with the cargo. This basic mechanism linked to the asymmetric distribution of the two nucleotide-bound forms of Ran between the nucleus and the cytoplasm generates a switch like mechanism controlling nucleo-cytoplasmic transport. Since 1999, we have known that after nuclear envelope breakdown (NEBD) Ran and the above transport receptors also provide a local control over the activity of factors driving spindle assembly and regulating other aspects of cell division. The identification and functional characterization of RanGTP mitotic targets is providing novel insights into mechanisms essential for cell division. Here we review our current knowledge on the RanGTP system and its regulation and we focus on the recent advances made through the characterization of its mitotic targets. We then briefly review the novel functions of the pathway that were recently described. Altogether, the RanGTP system has moonlighting functions exerting a spatial control over protein interactions that drive specific functions depending on the cellular context.
Collapse
Affiliation(s)
- Tommaso Cavazza
- Cell and Developmental Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain
| | - Isabelle Vernos
- Cell and Developmental Biology, Centre for Genomic Regulation, The Barcelona Institute of Science and TechnologyBarcelona, Spain; Universitat Pompeu FabraBarcelona, Spain; Institució Catalana de Recerca I Estudis AvançatsBarcelona, Spain
| |
Collapse
|
43
|
Menon S, Gupton SL. Building Blocks of Functioning Brain: Cytoskeletal Dynamics in Neuronal Development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 322:183-245. [PMID: 26940519 PMCID: PMC4809367 DOI: 10.1016/bs.ircmb.2015.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neural connectivity requires proper polarization of neurons, guidance to appropriate target locations, and establishment of synaptic connections. From when neurons are born to when they finally reach their synaptic partners, neurons undergo constant rearrangment of the cytoskeleton to achieve appropriate shape and polarity. Of particular importance to neuronal guidance to target locations is the growth cone at the tip of the axon. Growth-cone steering is also dictated by the underlying cytoskeleton. All these changes require spatiotemporal control of the cytoskeletal machinery. This review summarizes the proteins that are involved in modulating the actin and microtubule cytoskeleton during the various stages of neuronal development.
Collapse
Affiliation(s)
- Shalini Menon
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America
| | - Stephanie L Gupton
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, United States of America; Neuroscience Center and Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC, United States of America; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, United States of America.
| |
Collapse
|
44
|
Biased Brownian motion as a mechanism to facilitate nanometer-scale exploration of the microtubule plus end by a kinesin-8. Proc Natl Acad Sci U S A 2015; 112:E3826-35. [PMID: 26150501 DOI: 10.1073/pnas.1500272112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Kinesin-8s are plus-end-directed motors that negatively regulate microtubule (MT) length. Well-characterized members of this subfamily (Kip3, Kif18A) exhibit two important properties: (i) They are "ultraprocessive," a feature enabled by a second MT-binding site that tethers the motors to a MT track, and (ii) they dissociate infrequently from the plus end. Together, these characteristics combined with their plus-end motility cause Kip3 and Kif18A to enrich preferentially at the plus ends of long MTs, promoting MT catastrophes or pausing. Kif18B, an understudied human kinesin-8, also limits MT growth during mitosis. In contrast to Kif18A and Kip3, localization of Kif18B to plus ends relies on binding to the plus-end tracking protein EB1, making the relationship between its potential plus-end-directed motility and plus-end accumulation unclear. Using single-molecule assays, we show that Kif18B is only modestly processive and that the motor switches frequently between directed and diffusive modes of motility. Diffusion is promoted by the tail domain, which also contains a second MT-binding site that decreases the off rate of the motor from the MT lattice. In cells, Kif18B concentrates at the extreme tip of a subset of MTs, superseding EB1. Our data demonstrate that kinesin-8 motors use diverse design principles to target MT plus ends, which likely target them to the plus ends of distinct MT subpopulations in the mitotic spindle.
Collapse
|
45
|
Burgess SG, Peset I, Joseph N, Cavazza T, Vernos I, Pfuhl M, Gergely F, Bayliss R. Aurora-A-Dependent Control of TACC3 Influences the Rate of Mitotic Spindle Assembly. PLoS Genet 2015; 11:e1005345. [PMID: 26134678 PMCID: PMC4489650 DOI: 10.1371/journal.pgen.1005345] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/09/2015] [Indexed: 11/21/2022] Open
Abstract
The essential mammalian gene TACC3 is frequently mutated and amplified in cancers and its fusion products exhibit oncogenic activity in glioblastomas. TACC3 functions in mitotic spindle assembly and chromosome segregation. In particular, phosphorylation on S558 by the mitotic kinase, Aurora-A, promotes spindle recruitment of TACC3 and triggers the formation of a complex with ch-TOG-clathrin that crosslinks and stabilises kinetochore microtubules. Here we map the Aurora-A-binding interface in TACC3 and show that TACC3 potently activates Aurora-A through a domain centered on F525. Vertebrate cells carrying homozygous F525A mutation in the endogenous TACC3 loci exhibit defects in TACC3 function, namely perturbed localization, reduced phosphorylation and weakened interaction with clathrin. The most striking feature of the F525A cells however is a marked shortening of mitosis, at least in part due to rapid spindle assembly. F525A cells do not exhibit chromosome missegregation, indicating that they undergo fast yet apparently faithful mitosis. By contrast, mutating the phosphorylation site S558 to alanine in TACC3 causes aneuploidy without a significant change in mitotic duration. Our work has therefore defined a regulatory role for the Aurora-A-TACC3 interaction beyond the act of phosphorylation at S558. We propose that the regulatory relationship between Aurora-A and TACC3 enables the transition from the microtubule-polymerase activity of TACC3-ch-TOG to the microtubule-crosslinking activity of TACC3-ch-TOG-clathrin complexes as mitosis progresses. Aurora-A-dependent control of TACC3 could determine the balance between these activities, thereby influencing not only spindle length and stability but also the speed of spindle formation with vital consequences for chromosome alignment and segregation.
Collapse
Affiliation(s)
- Selena G. Burgess
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
- Cancer Research UK Leicester Centre, University of Leicester, Leicester, United Kingdom
| | - Isabel Peset
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Nimesh Joseph
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Tommaso Cavazza
- Cell and Developmental Biology program, Centre for Genomic Regulation (CRG), Barcelona, Spain
| | - Isabelle Vernos
- Cell and Developmental Biology program, Centre for Genomic Regulation (CRG), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Mark Pfuhl
- Cardiovascular and Randall Division, King’s College London, London, United Kingdom
| | - Fanni Gergely
- Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, United Kingdom
| | - Richard Bayliss
- Department of Biochemistry, University of Leicester, Leicester, United Kingdom
- Cancer Research UK Leicester Centre, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
46
|
Bearce EA, Erdogan B, Lowery LA. TIPsy tour guides: how microtubule plus-end tracking proteins (+TIPs) facilitate axon guidance. Front Cell Neurosci 2015; 9:241. [PMID: 26175669 PMCID: PMC4485311 DOI: 10.3389/fncel.2015.00241] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 06/15/2015] [Indexed: 01/01/2023] Open
Abstract
The growth cone is a dynamic cytoskeletal vehicle, which drives the end of a developing axon. It serves to interpret and navigate through the complex landscape and guidance cues of the early nervous system. The growth cone’s distinctive cytoskeletal organization offers a fascinating platform to study how extracellular cues can be translated into mechanical outgrowth and turning behaviors. While many studies of cell motility highlight the importance of actin networks in signaling, adhesion, and propulsion, both seminal and emerging works in the field have highlighted a unique and necessary role for microtubules (MTs) in growth cone navigation. Here, we focus on the role of singular pioneer MTs, which extend into the growth cone periphery and are regulated by a diverse family of microtubule plus-end tracking proteins (+TIPs). These +TIPs accumulate at the dynamic ends of MTs, where they are well-positioned to encounter and respond to key signaling events downstream of guidance receptors, catalyzing immediate changes in microtubule stability and actin cross-talk, that facilitate both axonal outgrowth and turning events.
Collapse
Affiliation(s)
| | - Burcu Erdogan
- Department of Biology, Boston College Chestnut Hill, MA, USA
| | | |
Collapse
|
47
|
Gutiérrez-Caballero C, Burgess SG, Bayliss R, Royle SJ. TACC3-ch-TOG track the growing tips of microtubules independently of clathrin and Aurora-A phosphorylation. Biol Open 2015; 4:170-9. [PMID: 25596274 PMCID: PMC4365485 DOI: 10.1242/bio.201410843] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 11/14/2014] [Indexed: 01/09/2023] Open
Abstract
The interaction between TACC3 (transforming acidic coiled coil protein 3) and the microtubule polymerase ch-TOG (colonic, hepatic tumor overexpressed gene) is evolutionarily conserved. Loading of TACC3-ch-TOG onto mitotic spindle microtubules requires the phosphorylation of TACC3 by Aurora-A kinase and the subsequent interaction of TACC3 with clathrin to form a microtubule-binding surface. Recent work indicates that TACC3 can track the plus-ends of microtubules and modulate microtubule dynamics in non-dividing cells via its interaction with ch-TOG. Whether there is a pool of TACC3-ch-TOG that is independent of clathrin in human cells, and what is the function of this pool, are open questions. Here, we describe the molecular interaction between TACC3 and ch-TOG that permits TACC3 recruitment to the plus-ends of microtubules. This TACC3-ch-TOG pool is independent of EB1, EB3, Aurora-A phosphorylation and binding to clathrin. We also describe the distinct combinatorial subcellular pools of TACC3, ch-TOG and clathrin. TACC3 is often described as a centrosomal protein, but we show that there is no significant population of TACC3 at centrosomes. The delineation of distinct protein pools reveals a simplified view of how these proteins are organized and controlled by post-translational modification.
Collapse
Affiliation(s)
| | - Selena G Burgess
- Cancer Research UK Leicester Centre and Department of Biochemistry, University of Leicester, Leicester LE1 9HN, UK
| | - Richard Bayliss
- Cancer Research UK Leicester Centre and Department of Biochemistry, University of Leicester, Leicester LE1 9HN, UK
| | - Stephen J Royle
- Division of Biomedical Cell Biology, Warwick Medical School, Gibbet Hill Road, Coventry, CV4 7AL, UK
| |
Collapse
|