1
|
Sasankan D, Mohan R. End Binding Proteins: Drivers of Cancer Progression. Cytoskeleton (Hoboken) 2024. [PMID: 39699076 DOI: 10.1002/cm.21972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Cancer, a complex and heterogeneous disease, continues to be a major global health concern. Despite advancements in diagnostics and therapeutics, the aggressive nature of certain cancers remain a significant challenge, necessitating a deeper understanding of the underlying molecular mechanisms driving their severity and progression. Cancer severity and progression depend on cellular properties such as cell migration, cell division, cell shape changes, and intracellular transport, all of which are driven by dynamic cellular microtubules. Dynamic properties of microtubules, in turn, are regulated by an array of proteins that influence their stability and growth. Among these regulators, End Binding (EB) proteins stand out as critical orchestrators of microtubule dynamics at their growing plus ends. Beyond their fundamental role in normal cellular functions, recent research has uncovered compelling evidence linking EB proteins to the pathogenesis of various diseases, including cancer progression. As the field of cancer research advances, the clinical implication of EB proteins role in cancer severity and aggressiveness become increasingly evident. This review aims to comprehensively explore the role of microtubule-associated EB proteins in influencing the severity and aggressiveness of cancer. We also discuss the potential significance of EB as a clinical biomarker for cancer diagnosis and prognosis and as a target for therapeutic intervention.
Collapse
Affiliation(s)
- Dhakshmi Sasankan
- Department of Biotechnology, University of Kerala, Kariavattom Campus, Thiruvananthapuram, India
| | - Renu Mohan
- Department of Biotechnology, University of Kerala, Kariavattom Campus, Thiruvananthapuram, India
| |
Collapse
|
2
|
Kim SK, Rogers SL, Lu W, Lee BS, Gelfand VI. EB-SUN, a new microtubule plus-end tracking protein in Drosophila. Mol Biol Cell 2024; 35:ar147. [PMID: 39475714 PMCID: PMC11656466 DOI: 10.1091/mbc.e24-09-0402] [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: 09/11/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
Microtubule (MT) regulation is essential for oocyte development. In Drosophila, MT stability, polarity, abundance, and orientation undergo dynamic changes across developmental stages. In our effort to identify novel microtubule-associated proteins that regulate MTs in the Drosophila ovary, we identified a previously uncharacterized gene, CG18190, which encodes a novel MT end-binding (EB) protein, which we propose to name EB-SUN. We show that EB-SUN colocalizes with EB1 at growing MT plus-ends in Drosophila S2 cells. Tissue-specific and developmental expression profiles from Paralog Explorer reveal that EB-SUN is predominantly expressed in the ovary and early embryos, while EB1 is ubiquitously expressed. Furthermore, as early as oocyte determination, EB-SUN comets are highly concentrated in oocytes during oogenesis. EB-SUN knockout (KO) results in decreased MT density at the onset of mid-oogenesis (stage 7) and delays oocyte growth during late mid-oogenesis (stage 9). Combining EB-SUN KO with EB1 knockdown (KD) in germ cells significantly further reduces MT density at stage 7. Hatching assays of single protein depletion reveal distinct roles for EB-SUN and EB1 in early embryogenesis, likely due to differences in their expression and binding partners. Notably, all eggs from EB-SUN KO/EB1 KD females fail to hatch, suggesting partial redundancy between these proteins.
Collapse
Affiliation(s)
- Sun K. Kim
- Department of Cell and Developmental Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611-3008
| | - Stephen L. Rogers
- Department of Biology, Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| | - Wen Lu
- Department of Cell and Developmental Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611-3008
| | - Brad S. Lee
- Department of Cell and Developmental Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611-3008
| | - Vladimir I. Gelfand
- Department of Cell and Developmental Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611-3008
| |
Collapse
|
3
|
Kim SK, Rogers SL, Lu W, Lee BS, Gelfand VI. EB-SUN, a New Microtubule Plus-End Tracking Protein in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612465. [PMID: 39314338 PMCID: PMC11419005 DOI: 10.1101/2024.09.11.612465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Microtubule (MT) regulation is essential for oocyte development. In Drosophila, MT stability, polarity, abundance, and orientation undergo dynamic changes across developmental stages. In our effort to identify novel microtubule-associated proteins (MAPs) that regulate MTs in the Drosophila ovary, we identified a previously uncharacterized gene, CG18190, encoding a novel MT end-binding (EB) protein, which we propose to name EB-SUN. We show that EB-SUN colocalizes with EB1 at growing microtubule plus-ends in Drosophila S2 cells. Tissue-specific and developmental expression profiles from Paralog Explorer reveal that EB-SUN is predominantly expressed in the ovary and early embryos, while EB1 is ubiquitously expressed. Furthermore, as early as oocyte determination, EB-SUN comets are highly concentrated in oocytes during oogenesis. EB-SUN knockout (KO) results in a decrease in MT density at the onset of mid-oogenesis (Stage 7) and delays oocyte growth during late mid-oogenesis (Stage 9). Combining EB-SUN KO with EB1 knockdown (KD) in germ cells significantly further reduced MT density at Stage 7. Notably, all eggs from EB-SUN KO/EB1 KD females fail to hatch, unlike single gene depletion, suggesting a functional redundancy between these two EB proteins during embryogenesis. Our findings indicate that EB-SUN and EB1 play distinct roles during early embryogenesis.
Collapse
Affiliation(s)
- Sun K Kim
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology Chicago, IL 60611-3008
| | - Stephen L Rogers
- Department of Biology, Integrative Program for Biological and Genome Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280
| | - Wen Lu
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology Chicago, IL 60611-3008
| | - Brad S Lee
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology Chicago, IL 60611-3008
| | - Vladimir I Gelfand
- Northwestern University, Feinberg School of Medicine, Department of Cell and Developmental Biology Chicago, IL 60611-3008
| |
Collapse
|
4
|
Radhakrishnan RM, Kizhakkeduth ST, Nair VM, Ayyappan S, Lakshmi RB, Babu N, Prasannajith A, Umeda K, Vijayan V, Kodera N, Manna TK. Kinetochore-microtubule attachment in human cells is regulated by the interaction of a conserved motif of Ska1 with EB1. J Biol Chem 2023; 299:102853. [PMID: 36592928 PMCID: PMC9926122 DOI: 10.1016/j.jbc.2022.102853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/16/2022] [Accepted: 12/18/2022] [Indexed: 01/02/2023] Open
Abstract
The kinetochore establishes the linkage between chromosomes and the spindle microtubule plus ends during mitosis. In vertebrates, the spindle-kinetochore-associated (Ska1,2,3) complex stabilizes kinetochore attachment with the microtubule plus ends, but how Ska is recruited to and stabilized at the kinetochore-microtubule interface is not understood. Here, our results show that interaction of Ska1 with the general microtubule plus end-associated protein EB1 through a conserved motif regulates Ska recruitment to kinetochores in human cells. Ska1 forms a stable complex with EB1 via interaction with the motif in its N-terminal disordered loop region. Disruption of this interaction either by deleting or mutating the motif disrupts Ska complex recruitment to kinetochores and induces chromosome alignment defects, but it does not affect Ska complex assembly. Atomic-force microscopy imaging revealed that Ska1 is anchored to the C-terminal region of the EB1 dimer through its loop and thereby promotes formation of extended structures. Furthermore, our NMR data showed that the Ska1 motif binds to the residues in EB1 that are the binding sites of other plus end targeting proteins that are recruited to microtubules by EB1 through a similar conserved motif. Collectively, our results demonstrate that EB1-mediated Ska1 recruitment onto the microtubule serves as a general mechanism for the formation of vertebrate kinetochore-microtubule attachments and metaphase chromosome alignment.
Collapse
Affiliation(s)
- Renjith M Radhakrishnan
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Safwa T Kizhakkeduth
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Vishnu M Nair
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Shine Ayyappan
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - R Bhagya Lakshmi
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Neethu Babu
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Anjaly Prasannajith
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Kenichi Umeda
- Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Vinesh Vijayan
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Noriyuki Kodera
- Nano Life Science Institute (WPI-Nano LSI), Kanazawa University, Kakuma-machi, Kanazawa, Japan
| | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
5
|
MAPRE2 regulates the first meiotic progression in mouse oocytes. Exp Cell Res 2022; 416:113135. [DOI: 10.1016/j.yexcr.2022.113135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 11/22/2022]
|
6
|
Ayyappan S, Dharan PS, Krishnan A, Marira RR, Lambert M, Manna TK, Vijayan V. SxIP binding disrupts the constitutive homodimer interface of EB1 and stabilizes EB1 monomer. Biophys J 2021; 120:2019-2029. [PMID: 33737159 DOI: 10.1016/j.bpj.2021.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/16/2021] [Accepted: 03/01/2021] [Indexed: 11/16/2022] Open
Abstract
SxIP is a microtubule tip localizing signal found in many +TIP proteins that bind to the hydrophobic cavity of the C-terminal domain of end binding protein 1 (EB1) and then positively regulate the microtubule plus-end tracking of EBs. However, the exact mechanism of microtubule activation of EBs in the presence of SxIP signaling motif is not known. Here, we studied the effect of SxIP peptide on the native conformation of EB1 in solution. Using various NMR experiments, we found that SxIP peptide promoted the dissociation of natively formed EB1 dimer. We also discovered that I224A mutation of EB1 resulted in an unfolded C-terminal domain, which upon binding with the SxIP motif folded to its native structure. Molecular dynamics simulations also confirmed the relative structural stability of EB1 monomer in the SxIP bound state. Residual dipolar couplings and heteronuclear NOE analysis suggested that the binding of SxIP peptide at the C-terminal domain of EB1 decreased the dynamics and conformational flexibility of the N-terminal domain involved in EB1-microtubule interaction. The SxIP-induced disruption of the dimeric interactions in EB1, coupled with the reduction in conformational flexibility of the N-terminal domain of EB1, might facilitate the microtubule association of EB1.
Collapse
Affiliation(s)
- Shine Ayyappan
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Pooja S Dharan
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Arya Krishnan
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Renjith R Marira
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Mahil Lambert
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India
| | - Vinesh Vijayan
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
7
|
Abdelkarim H, Hitchinson B, Qu X, Banerjee A, Komarova YA, Gaponenko V. NMR resonance assignment and structure prediction of the C-terminal domain of the microtubule end-binding protein 3. PLoS One 2020; 15:e0232338. [PMID: 32421702 PMCID: PMC7233555 DOI: 10.1371/journal.pone.0232338] [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: 10/25/2019] [Accepted: 04/13/2020] [Indexed: 11/29/2022] Open
Abstract
End-binding proteins (EBs) associate with the growing microtubule plus ends to regulate microtubule dynamics as well as the interaction with intracellular structures. EB3 contributes to pathological vascular leakage through interacting with the inositol 1,4,5-trisphosphate receptor 3 (IP3R3), a calcium channel located at the endoplasmic reticulum membrane. The C-terminal domain of EB3 (residues 200–281) is functionally important for this interaction because it contains the effector binding sites, a prerequisite for EB3 activity and specificity. Structural data for this domain is limited. Here, we report the backbone chemical shift assignments for the human EB3 C-terminal domain and computationally explore its EB3 conformations. Backbone assignments, along with computational models, will allow future investigation of EB3 structural dynamics, interactions with effectors, and will facilitate the development of novel EB3 inhibitors.
Collapse
Affiliation(s)
- Hazem Abdelkarim
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Ben Hitchinson
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Xinyan Qu
- Department of Pharmacology and the Center for Lung Biology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Avik Banerjee
- Department of Chemistry, University of Illinois, Chicago, IL, United States of America
| | - Yulia A. Komarova
- Department of Pharmacology and the Center for Lung Biology, University of Illinois at Chicago, Chicago, IL, United States of America
- * E-mail: (YAK); (VG)
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States of America
- * E-mail: (YAK); (VG)
| |
Collapse
|
8
|
Kruse K, Klomp J, Sun M, Chen Z, Santana D, Huang F, Kanabar P, Maienschein-Cline M, Komarova YA. Analysis of biological networks in the endothelium with biomimetic microsystem platform. Am J Physiol Lung Cell Mol Physiol 2019; 317:L392-L401. [PMID: 31313617 DOI: 10.1152/ajplung.00392.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Here we describe a novel method for studying the protein "interactome" in primary human cells and apply this method to investigate the effect of posttranslational protein modifications (PTMs) on the protein's functions. We created a novel "biomimetic microsystem platform" (Bio-MSP) to isolate the protein complexes in primary cells by covalently attaching purified His-tagged proteins to a solid microscale support. Using this Bio-MSP, we have analyzed the interactomes of unphosphorylated and phosphomimetic end-binding protein-3 (EB3) in endothelial cells. Pathway analysis of these interactomes demonstrated the novel role of EB3 phosphorylation at serine 162 in regulating the protein's function. We showed that phosphorylation "switches" the EB3 biological network to modulate cellular processes such as cell-to-cell adhesion whereas dephosphorylation of this site promotes cell proliferation. This novel technique provides a useful tool to study the role of PTMs or single point mutations in activating distinct signal transduction networks and thereby the biological function of the protein in health and disease.
Collapse
Affiliation(s)
- Kevin Kruse
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Jeff Klomp
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Mitchell Sun
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Zhang Chen
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Dianicha Santana
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Fei Huang
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Pinal Kanabar
- Research Informatics Core of the Research Resources Center, University of Illinois at Chicago, Chicago, Illinois.,College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Mark Maienschein-Cline
- Research Informatics Core of the Research Resources Center, University of Illinois at Chicago, Chicago, Illinois.,College of Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - Yulia A Komarova
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
9
|
Blanco C, Morales D, Mogollones I, Vergara‐Jaque A, Vargas C, Álvarez A, Riquelme D, Leiva‐Salcedo E, González W, Morales D, Maureira D, Aldunate I, Cáceres M, Varela D, Cerda O. EB1‐ and EB2‐dependent anterograde trafficking of TRPM4 regulates focal adhesion turnover and cell invasion. FASEB J 2019; 33:9434-9452. [DOI: 10.1096/fj.201900136r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Constanza Blanco
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Danna Morales
- Program of Physiology and Biophysics Institute of Biomedical Sciences Faculty of Medicine Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Ignacio Mogollones
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Ariela Vergara‐Jaque
- Program of Physiology and Biophysics Institute of Biomedical Sciences Faculty of Medicine Universidad de Chile Santiago Chile
- Multidisciplinary Scientific Nucleus Universidad de Talca Talca Chile
- Center for Bioinformatics and Molecular Simulation Universidad de Talca Talca Chile
| | - Carla Vargas
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Alhejandra Álvarez
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Denise Riquelme
- Department of Biology Faculty of Chemistry and Biology Universidad de Santiago de Chile Santiago Chile
| | - Elías Leiva‐Salcedo
- Department of Biology Faculty of Chemistry and Biology Universidad de Santiago de Chile Santiago Chile
| | - Wendy González
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
- Center for Bioinformatics and Molecular Simulation Universidad de Talca Talca Chile
| | - Diego Morales
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Diego Maureira
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Ismael Aldunate
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
| | - Mónica Cáceres
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
- The Wound Repair Treatment, and Health (WoRTH) Initiative Santiago Chile
| | - Diego Varela
- Program of Physiology and Biophysics Institute of Biomedical Sciences Faculty of Medicine Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
| | - Oscar Cerda
- Program of Cellular and Molecular Biology Universidad de Chile Santiago Chile
- Millennium Nucleus of Ion Channels‐Associated Diseases (MiNICAD) Santiago Chile
- The Wound Repair Treatment, and Health (WoRTH) Initiative Santiago Chile
| |
Collapse
|
10
|
Mal3 is a multi-copy suppressor of the sensitivity to microtubule-depolymerizing drugs and chromosome mis-segregation in a fission yeast pka1 mutant. PLoS One 2019; 14:e0214803. [PMID: 30973898 PMCID: PMC6459531 DOI: 10.1371/journal.pone.0214803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 03/20/2019] [Indexed: 11/24/2022] Open
Abstract
The cAMP-dependent protein kinase Pka1 is known as a regulator of glycogenesis, transition into meiosis, chronological aging, and stress responses in the fission yeast, Schizosaccharomyces pombe. We demonstrated here that Pka1 is responsible for normal growth in the presence of the microtubule-destabilization drug TBZ and proper chromosome segregation. The deletion of the pka1 gene resulted in the TBZ-sensitive phenotype and chromosome mis-segregation. We isolated the mal3 gene as a multi-copy suppressor of the TBZ-sensitive phenotype in the pka1Δ strains. Overexpression of the CH domain (1–143) or the high-affinity microtubule binding mutant (1–143 Q89R) of Mal3 rescued the TBZ-sensitive phenotype in the pka1Δ and mal3Δ strains, while the EB1 domain (135–308) and the mutants defective in microtubule binding (1–143 Q89E) failed to do so in the same strains. Chromosome mis-segregation caused by TBZ in the pka1Δ or mal3Δ strains was suppressed by the overexpression of the Mal3 CH domain (1–143), Mal3 CH domain with the coiled-coil domain (1–197), or full-length Mal3. Overexpression of EB1 orthologs from Saccharomyces cerevisiae, Arabidopsis thaliana, Mus musculus, or Homo sapiens suppressed the TBZ-sensitive phenotype in the pka1Δ strains, indicating their conserved functions. These findings suggest that Pka1 and the microtubule binding of the Mal3 CH domain play a role in the maintenance of proper chromosome segregation.
Collapse
|
11
|
van Haren J, Wittmann T. Microtubule Plus End Dynamics - Do We Know How Microtubules Grow?: Cells boost microtubule growth by promoting distinct structural transitions at growing microtubule ends. Bioessays 2019; 41:e1800194. [PMID: 30730055 PMCID: PMC7021488 DOI: 10.1002/bies.201800194] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/22/2018] [Indexed: 12/31/2022]
Abstract
Microtubules form a highly dynamic filament network in all eukaryotic cells. Individual microtubules grow by tubulin dimer subunit addition and frequently switch between phases of growth and shortening. These unique dynamics are powered by GTP hydrolysis and drive microtubule network remodeling, which is central to eukaryotic cell biology and morphogenesis. Yet, our knowledge of the molecular events at growing microtubule ends remains incomplete. Here, recent ultrastructural, biochemical and cell biological data are integrated to develop a realistic model of growing microtubule ends comprised of structurally distinct but biochemically overlapping zones. Proteins that recognize microtubule lattice conformations associated with specific tubulin guanosine nucleotide states may independently control major structural transitions at growing microtubule ends. A model is proposed in which tubulin dimer addition and subsequent closure of the MT wall are optimized in cells to achieve rapid physiological microtubule growth.
Collapse
Affiliation(s)
- Jeffrey van Haren
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
12
|
Chen Y, Wang P, Slep KC. Mapping multivalency in the CLIP-170-EB1 microtubule plus-end complex. J Biol Chem 2018; 294:918-931. [PMID: 30455356 DOI: 10.1074/jbc.ra118.006125] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/09/2018] [Indexed: 11/06/2022] Open
Abstract
Cytoplasmic linker protein 170 (CLIP-170) is a microtubule plus-end factor that links vesicles to microtubules and recruits the dynein-dynactin complex to microtubule plus ends. CLIP-170 plus-end localization is end binding 1 (EB1)-dependent. CLIP-170 contains two N-terminal cytoskeleton-associated protein glycine-rich (CAP-Gly) domains flanked by serine-rich regions. The CAP-Gly domains are known EB1-binding domains, and the serine-rich regions have also been implicated in CLIP-170's microtubule plus-end localization mechanism. However, the determinants in these serine-rich regions have not been identified. Here we elucidated multiple EB1-binding modules in the CLIP-170 N-terminal region. Using isothermal titration calorimetry and size-exclusion chromatography, we mapped and biophysically characterized these EB1-binding modules, including the two CAP-Gly domains, a bridging SXIP motif, and a unique array of divergent SXIP-like motifs located N-terminally to the first CAP-Gly domain. We found that, unlike the EB1-binding mode of the CAP-Gly domain in the dynactin-associated protein p150Glued, which dually engages the EB1 C-terminal EEY motif as well as the EB homology domain and sterically occludes SXIP motif binding, the CLIP-170 CAP-Gly domains engage only the EEY motif, enabling the flanking SXIP and SXIP-like motifs to bind the EB homology domain. These multivalent EB1-binding modules provided avidity to the CLIP-170-EB1 interaction, likely clarifying why CLIP-170 preferentially binds EB1 rather than the α-tubulin C-terminal EEY motif. Our finding that CLIP-170 has multiple non-CAP-Gly EB1-binding modules may explain why autoinhibition of CLIP-170 GAP-Gly domains does not fully abrogate its microtubule plus-end localization. This work expands our understanding of EB1-binding motifs and their multivalent networks.
Collapse
Affiliation(s)
- Yaodong Chen
- From the Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, Shaanxi 710069, China.,the Department of Biology, University of North Carolina, Chapel Hill, North Carolina 27599-3280, and
| | - Ping Wang
- the Department of Neurology, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Kevin C Slep
- the Department of Biology, University of North Carolina, Chapel Hill, North Carolina 27599-3280, and
| |
Collapse
|
13
|
Gireesh KK, Shine A, Lakshmi RB, Vijayan V, Manna TK. GTP-binding facilitates EB1 recruitment onto microtubules by relieving its auto-inhibition. Sci Rep 2018; 8:9792. [PMID: 29955158 PMCID: PMC6023887 DOI: 10.1038/s41598-018-28056-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 06/14/2018] [Indexed: 12/23/2022] Open
Abstract
Microtubule plus end-binding protein, EB1 is a key regulator of microtubule dynamics. Auto-inhibitory interaction in EB1 has previously been shown to inhibit its ability to bind to microtubules and regulate microtubule dynamics. However, the factors that promote its microtubule regulatory activity by over-coming the auto-inhibition are less known. Here, we show that GTP plays a critical role in promoting the microtubule-targeting activity of EB1 by suppressing its auto-inhibition. Our biophysical data demonstrate that GTP binds to EB1 at a distinct site in its conserved N-terminal domain. Detailed analyses reveal that GTP-binding suppresses the intra-molecular inhibitory interaction between the globular N-terminus and the C-terminal coiled-coil domain. We further show that mutation of the GTP-binding site residues in N-terminus weakens the affinity for GTP, but also for the C-terminus, indicating overlapping binding sites. Confocal imaging and biochemical analysis reveal that EB1 localization on the microtubules is significantly increased upon mutations of the GTP-binding site residues. The results demonstrate a unique role of GTP in facilitating EB1 interaction with the microtubules by relieving its intra-molecular inhibition. They also implicate that GTP-binding may regulate the functions of EB1 on the cellular microtubules.
Collapse
Affiliation(s)
- K K Gireesh
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, CET Campus, Thiruvananthapuram, 695016, Kerala, India
| | - A Shine
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, CET Campus, Thiruvananthapuram, 695016, Kerala, India
| | - R Bhagya Lakshmi
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, CET Campus, Thiruvananthapuram, 695016, Kerala, India
| | - Vinesh Vijayan
- School of Chemistry, Indian Institute of Science Education and Research Thiruvananthapuram, CET Campus, Thiruvananthapuram, 695016, Kerala, India.
| | - Tapas K Manna
- School of Biology, Indian Institute of Science Education and Research Thiruvananthapuram, CET Campus, Thiruvananthapuram, 695016, Kerala, India.
| |
Collapse
|
14
|
Procter DJ, Banerjee A, Nukui M, Kruse K, Gaponenko V, Murphy EA, Komarova Y, Walsh D. The HCMV Assembly Compartment Is a Dynamic Golgi-Derived MTOC that Controls Nuclear Rotation and Virus Spread. Dev Cell 2018; 45:83-100.e7. [PMID: 29634939 DOI: 10.1016/j.devcel.2018.03.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 02/08/2018] [Accepted: 03/14/2018] [Indexed: 10/17/2022]
Abstract
Human cytomegalovirus (HCMV), a leading cause of congenital birth defects, forms an unusual cytoplasmic virion maturation site termed the "assembly compartment" (AC). Here, we show that the AC also acts as a microtubule-organizing center (MTOC) wherein centrosome activity is suppressed and Golgi-based microtubule (MT) nucleation is enhanced. This involved viral manipulation of discrete functions of MT plus-end-binding (EB) proteins. In particular, EB3, but not EB1 or EB2, was recruited to the AC and was required to nucleate MTs that were rapidly acetylated. EB3-regulated acetylated MTs were necessary for nuclear rotation prior to cell migration, maintenance of AC structure, and optimal virus replication. Independently, a myristoylated peptide that blocked EB3-mediated enrichment of MT regulatory proteins at Golgi regions of the AC also suppressed acetylated MT formation, nuclear rotation, and infection. Thus, HCMV offers new insights into the regulation and functions of Golgi-derived MTs and the therapeutic potential of targeting EB3.
Collapse
Affiliation(s)
- Dean J Procter
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Avik Banerjee
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Masatoshi Nukui
- Department of Translational Medicine, Baruch S. Blumberg Research Institute, Doylestown, PA 18902, USA; Forge Life Science, Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA
| | - Kevin Kruse
- Department of Pharmacology and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Eain A Murphy
- Department of Translational Medicine, Baruch S. Blumberg Research Institute, Doylestown, PA 18902, USA; Forge Life Science, Pennsylvania Biotechnology Center, Doylestown, PA 18902, USA
| | - Yulia Komarova
- Department of Pharmacology and The Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Derek Walsh
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
15
|
van Haren J, Charafeddine RA, Ettinger A, Wang H, Hahn KM, Wittmann T. Local control of intracellular microtubule dynamics by EB1 photodissociation. Nat Cell Biol 2018; 20:252-261. [PMID: 29379139 PMCID: PMC5826794 DOI: 10.1038/s41556-017-0028-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 12/13/2017] [Indexed: 12/18/2022]
Abstract
End-binding proteins, EBs, are adaptors that recruit functionally diverse +TIP proteins to growing microtubule plus ends. To test with high spatial and temporal accuracy how, when and where +TIP protein complexes contribute to dynamic cell biology, we developed a photo-inactivated EB1 variant (π-EB1) by inserting a blue light-sensitive protein-protein interaction module between the microtubule- and +TIP-binding domains of EB1. π-EB1 replaces endogenous EB1 function in the absence of blue light. In contrast, blue light-mediated π-EB1 photo-dissociation results in rapid +TIP complex disassembly, and acutely and reversibly attenuates microtubule growth independent of microtubule end association of the microtubule polymerase CKAP5 (ch-TOG, XMAP215). Local π-EB1 photo-dissociation allows subcellular microtubule dynamics control at the second and micrometre scale, and elicits aversive turning of migrating cancer cells. Importantly, light-mediated domain splitting can serve as template to optically control other intracellular protein activities.
Collapse
Affiliation(s)
- Jeffrey van Haren
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Rabab A Charafeddine
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Andreas Ettinger
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA.,Institute of Epigenetics and Stem Cells, Helmholtz Center Munich, München, Germany
| | - Hui Wang
- University of North Carolina, Chapel Hill, NC, USA
| | - Klaus M Hahn
- University of North Carolina, Chapel Hill, NC, USA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA.
| |
Collapse
|
16
|
Targeting SxIP-EB1 interaction: An integrated approach to the discovery of small molecule modulators of dynamic binding sites. Sci Rep 2017; 7:15533. [PMID: 29138501 PMCID: PMC5686072 DOI: 10.1038/s41598-017-15502-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/25/2017] [Indexed: 11/09/2022] Open
Abstract
End binding protein 1 (EB1) is a key element in the complex network of protein-protein interactions at microtubule (MT) growing ends, which has a fundamental role in MT polymerisation. EB1 is an important protein target as it is involved in regulating MT dynamic behaviour, and has been associated with several disease states, such as cancer and neuronal diseases. Diverse EB1 binding partners are recognised through a conserved four amino acid motif, (serine-X-isoleucine-proline) which exists within an intrinsically disordered region. Here we report the use of a multidisciplinary computational and experimental approach for the discovery of the first small molecule scaffold which targets the EB1 recruiting domain. This approach includes virtual screening (structure- and ligand-based design) and multiparameter compound selection. Subsequent studies on the selected compounds enabled the elucidation of the NMR structures of the C-terminal domain of EB1 in the free form and complexed with a small molecule. These structures show that the binding site is not preformed in solution, and ligand binding is fundamental for the binding site formation. This work is a successful demonstration of the combination of modelling and experimental methods to enable the discovery of compounds which bind to these challenging systems.
Collapse
|
17
|
Ivashko-Pachima Y, Sayas CL, Malishkevich A, Gozes I. ADNP/NAP dramatically increase microtubule end-binding protein-Tau interaction: a novel avenue for protection against tauopathy. Mol Psychiatry 2017; 22:1335-1344. [PMID: 28115743 DOI: 10.1038/mp.2016.255] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/20/2016] [Accepted: 12/06/2016] [Indexed: 12/30/2022]
Abstract
Activity-dependent neuroprotective protein (ADNP), vital for brain formation and cognitive function, is mutated in autism and linked to neurodegenerative/psychiatric diseases. An eight-amino-acid peptide snippet of ADNP, NAP (NAPVSIPQ), identified as a smallest active fragment, includes the SxIP microtubule (MT) end-binding protein (EB) association motif, and enhances ADNP-EB3 interaction. Depletion of EB1 or EB3 abolishes NAP protection against zinc intoxication. Furthermore, NAP enhances Tau-MT interaction, and Tau regulates the localization and function of EB1 and EB3 in developing neuronal cells. Here, we asked how NAP (ADNP) enhances Tau-MT interactions and whether this is mediated by EBs. We showed, for we believe the first time, that NAP augmented endogenous EB1 comet density in the N1E-115 neuroblastoma neuronal model. This finding was substantiated by cell transfection with fluorescent EB1 and live cell imaging. NAP increased comet amounts, length and speed. At the molecular level, NAP enhanced EB3 homodimer formation, while decreasing EB1-EB3 heterodimer content and driving EB1- and EB3-Tau interactions (dramatic 20-fold increases), leading to recruitment of EB1/EB3 and Tau to MTs under zinc intoxication. Our previous results showed that while NAP protected neuronal-like cells against oxidative stress, it did not protect NIH3T3 fibroblasts. Here, NAP did not protect NIH3T3 cells against zinc intoxication, unless these cells were transfected with Tau. Interestingly, other MT associated proteins (MAPs) may replace Tau, thus, EB-Tau (MAPs) interaction is identified as a novel target for endogenous ADNP neuroprotection, and a future target for drug development, with NAP as a prototype.
Collapse
Affiliation(s)
- Y Ivashko-Pachima
- The Lily and Avraham Gildor Chair for the Investigation of Growth Factors, Dr. Diana and Zelman Elton (Elbaum) Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - C Laura Sayas
- Centre for Biomedical Research of the Canary Islands (CIBICAN), Institute for Biomedical Technologies (ITB), University of La Laguna (ULL), Tenerife, Spain
| | - A Malishkevich
- The Lily and Avraham Gildor Chair for the Investigation of Growth Factors, Dr. Diana and Zelman Elton (Elbaum) Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| | - I Gozes
- The Lily and Avraham Gildor Chair for the Investigation of Growth Factors, Dr. Diana and Zelman Elton (Elbaum) Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neuroscience and Adams Super Center for Brain Studies, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
18
|
Mustyatsa VV, Boyakhchyan AV, Ataullakhanov FI, Gudimchuk NB. EB-family proteins: Functions and microtubule interaction mechanisms. BIOCHEMISTRY (MOSCOW) 2017; 82:791-802. [DOI: 10.1134/s0006297917070045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Nehlig A, Molina A, Rodrigues-Ferreira S, Honoré S, Nahmias C. Regulation of end-binding protein EB1 in the control of microtubule dynamics. Cell Mol Life Sci 2017; 74:2381-2393. [PMID: 28204846 PMCID: PMC11107513 DOI: 10.1007/s00018-017-2476-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/13/2017] [Accepted: 01/24/2017] [Indexed: 12/14/2022]
Abstract
The regulation of microtubule dynamics is critical to ensure essential cell functions, such as proper segregation of chromosomes during mitosis or cell polarity and migration. End-binding protein 1 (EB1) is a plus-end-tracking protein (+TIP) that accumulates at growing microtubule ends and plays a pivotal role in the regulation of microtubule dynamics. EB1 autonomously binds an extended tubulin-GTP/GDP-Pi structure at growing microtubule ends and acts as a molecular scaffold that recruits a large number of regulatory +TIPs through interaction with CAP-Gly or SxIP motifs. While extensive studies have focused on the structure of EB1-interacting site at microtubule ends and its role as a molecular platform, the mechanisms involved in the negative regulation of EB1 have only started to emerge and remain poorly understood. In this review, we summarize recent studies showing that EB1 association with MT ends is regulated by post-translational modifications and affected by microtubule-targeting agents. We also present recent findings that structural MAPs, that have no tip-tracking activity, physically interact with EB1 to prevent its accumulation at microtubule plus ends. These observations point out a novel concept of "endogenous EB1 antagonists" and emphasize the importance of finely regulating EB1 function at growing microtubule ends.
Collapse
Affiliation(s)
- Anne Nehlig
- Inserm U981, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
- University Paris Saclay, 94800, Villejuif, France
| | - Angie Molina
- Inserm U981, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
- University Paris Saclay, 94800, Villejuif, France
- CBD, University of Toulouse-3, Toulouse, France
| | - Sylvie Rodrigues-Ferreira
- Inserm U981, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France
- University Paris Saclay, 94800, Villejuif, France
| | - Stéphane Honoré
- Aix Marseille University, Inserm U-911, CRO2, Marseille, France
- Service Pharmacie, CHU Hôpital de La Timone, APHM, Marseille, France
| | - Clara Nahmias
- Inserm U981, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94800, Villejuif, France.
- University Paris Saclay, 94800, Villejuif, France.
| |
Collapse
|
20
|
Ayanlaja AA, Xiong Y, Gao Y, Ji G, Tang C, Abdikani Abdullah Z, Gao D. Distinct Features of Doublecortin as a Marker of Neuronal Migration and Its Implications in Cancer Cell Mobility. Front Mol Neurosci 2017; 10:199. [PMID: 28701917 PMCID: PMC5487455 DOI: 10.3389/fnmol.2017.00199] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/06/2017] [Indexed: 12/16/2022] Open
Abstract
Neuronal migration is a critical process in the development of the nervous system. Defects in the migration of the neurons are associated with diseases like lissencephaly, subcortical band heterotopia (SBH), and pachygyria. Doublecortin (DCX) is an essential factor in neurogenesis and mutations in this protein impairs neuronal migration leading to several pathological conditions. Although, DCX is capable of modulating and stabilizing microtubules (MTs) to ensure effective migration, the mechanisms involved in executing these functions remain poorly understood. Meanwhile, there are existing gaps regarding the processes that underlie tumor initiation and progression into cancer as well as the ability to migrate and invade normal cells. Several studies suggest that DCX is involved in cancer metastasis. Unstable interactions between DCX and MTs destabilizes cytoskeletal organization leading to disorganized movements of cells, a process which may be implicated in the uncontrolled migration of cancer cells. However, the underlying mechanism is complex and require further clarification. Therefore, exploring the importance and features known up to date about this molecule will broaden our understanding and shed light on potential therapeutic approaches for the associated neurological diseases. This review summarizes current knowledge about DCX, its features, functions, and relationships with other proteins. We also present an overview of its role in cancer cells and highlight the importance of studying its gene mutations.
Collapse
Affiliation(s)
- Abiola A Ayanlaja
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - Ye Xiong
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - Yue Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - GuangQuan Ji
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - Zamzam Abdikani Abdullah
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| | - DianShuai Gao
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical UniversityXuzhou, China
| |
Collapse
|
21
|
Fong KW, Au FKC, Jia Y, Yang S, Zhou L, Qi RZ. Microtubule plus-end tracking of end-binding protein 1 (EB1) is regulated by CDK5 regulatory subunit-associated protein 2. J Biol Chem 2017; 292:7675-7687. [PMID: 28320860 DOI: 10.1074/jbc.m116.759746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 03/15/2017] [Indexed: 01/08/2023] Open
Abstract
Microtubules are polar cytoskeleton filaments that extend via growth at their plus ends. Microtubule plus-end-tracking proteins (+TIPs) accumulate at these growing plus ends to control microtubule dynamics and attachment. The +TIP end-binding protein 1 (EB1) and its homologs possess an autonomous plus-end-tracking mechanism and interact with other known +TIPs, which then recruit those +TIPs to the growing plus ends. A major +TIP class contains the SXIP (Ser-X-Ile-Pro, with X denoting any amino acid residue) motif, known to interact with EB1 and its homologs for plus-end tracking, but the role of SXIP in regulating EB1 activities is unclear. We show here that an interaction of EB1 with the SXIP-containing +TIP CDK5 regulatory subunit-associated protein 2 (CDK5RAP2) regulates several EB1 activities, including microtubule plus-end tracking, dynamics at microtubule plus ends, microtubule and α/β-tubulin binding, and microtubule polymerization. The SXIP motif fused with a dimerization domain from CDK5RAP2 significantly enhanced EB1 plus-end-tracking and microtubule-polymerizing and bundling activities, but the SXIP motif alone failed to do so. An SXIP-binding-deficient EB1 mutant displayed significantly lower microtubule plus-end tracking than the wild-type protein in transfected cells. These results suggest that EB1 cooperates with CDK5RAP2 and perhaps other SXIP-containing +TIPs in tracking growing microtubule tips. We also generated plus-end-tracking chimeras of CDK5RAP2 and the adenomatous polyposis coli protein (APC) and found that overexpression of the dimerization domains interfered with microtubule plus-end tracking of their respective SXIP-containing chimeras. Our results suggest that disruption of SXIP dimerization enables detailed investigations of microtubule plus-end-associated functions of individual SXIP-containing +TIPs.
Collapse
Affiliation(s)
- Ka-Wing Fong
- From the Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Franco K C Au
- From the Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yue Jia
- From the Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Shaozhong Yang
- From the Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Liying Zhou
- From the Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Robert Z Qi
- From the Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| |
Collapse
|
22
|
EB1 contributes to proper front-to-back polarity in neutrophil-like HL-60 cells. Eur J Cell Biol 2017; 96:143-153. [DOI: 10.1016/j.ejcb.2017.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 12/10/2016] [Accepted: 01/16/2017] [Indexed: 12/30/2022] Open
|
23
|
Ramirez-Rios S, Serre L, Stoppin-Mellet V, Prezel E, Vinit A, Courriol E, Fourest-Lieuvin A, Delaroche J, Denarier E, Arnal I. A TIRF microscopy assay to decode how tau regulates EB’s tracking at microtubule ends. Methods Cell Biol 2017; 141:179-197. [DOI: 10.1016/bs.mcb.2017.06.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
24
|
Matsuo Y, Maurer SP, Yukawa M, Zakian S, Singleton MR, Surrey T, Toda T. An unconventional interaction between Dis1/TOG and Mal3/EB1 in fission yeast promotes the fidelity of chromosome segregation. J Cell Sci 2016; 129:4592-4606. [PMID: 27872152 PMCID: PMC5201023 DOI: 10.1242/jcs.197533] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 11/09/2016] [Indexed: 12/31/2022] Open
Abstract
Dynamic microtubule plus-ends interact with various intracellular target regions such as the cell cortex and the kinetochore. Two conserved families of microtubule plus-end-tracking proteins, the XMAP215, ch-TOG or CKAP5 family and the end-binding 1 (EB1, also known as MAPRE1) family, play pivotal roles in regulating microtubule dynamics. Here, we study the functional interplay between fission yeast Dis1, a member of the XMAP215/TOG family, and Mal3, an EB1 protein. Using an in vitro microscopy assay, we find that purified Dis1 autonomously tracks growing microtubule ends and is a bona fide microtubule polymerase. Mal3 recruits additional Dis1 to microtubule ends, explaining the synergistic enhancement of microtubule dynamicity by these proteins. A non-canonical binding motif in Dis1 mediates the interaction with Mal3. X-ray crystallography shows that this new motif interacts in an unconventional configuration with the conserved hydrophobic cavity formed within the Mal3 C-terminal region that typically interacts with the canonical SXIP motif. Selectively perturbing the Mal3-Dis1 interaction in living cells demonstrates that it is important for accurate chromosome segregation. Whereas, in some metazoans, the interaction between EB1 and the XMAP215/TOG family members requires an additional binding partner, fission yeast relies on a direct interaction, indicating evolutionary plasticity of this critical interaction module.
Collapse
Affiliation(s)
- Yuzy Matsuo
- Synthetic and Systems Biochemistry of the Microtubule Cytoskeleton Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Cell Regulation Laboratory, The Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Sebastian P Maurer
- Synthetic and Systems Biochemistry of the Microtubule Cytoskeleton Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Cell and Developmental Biology, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona 08002, Spain
| | - Masashi Yukawa
- Hiroshima Research Center for Healthy Aging (HiHA), Department of Molecular Biotechnology, Graduate School of Advanced Science of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8530, Japan
| | - Silva Zakian
- Structural Biology of Chromosome Segregation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Martin R Singleton
- Structural Biology of Chromosome Segregation Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Thomas Surrey
- Synthetic and Systems Biochemistry of the Microtubule Cytoskeleton Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Takashi Toda
- Cell Regulation Laboratory, The Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
- Hiroshima Research Center for Healthy Aging (HiHA), Department of Molecular Biotechnology, Graduate School of Advanced Science of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8530, Japan
| |
Collapse
|
25
|
Kim J, Lee HY, Lee KH, Park SJ. Phosphorylation of Serine 148 in Giardia lamblia
End-binding 1 Protein is Important for Cell Division. J Eukaryot Microbiol 2016; 64:464-480. [DOI: 10.1111/jeu.12384] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 11/07/2016] [Indexed: 01/10/2023]
Affiliation(s)
- Juri Kim
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine; Seoul 03722 South Korea
| | - Hye-Yeon Lee
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine; Seoul 03722 South Korea
| | - Kyu-Ho Lee
- Department of Life Science; Sogang University; Seoul 04107 South Korea
| | - Soon-Jung Park
- Department of Environmental Medical Biology and Institute of Tropical Medicine, Brain Korea 21 PLUS Project for Medical Science; Yonsei University College of Medicine; Seoul 03722 South Korea
| |
Collapse
|
26
|
Ramirez-Rios S, Denarier E, Prezel E, Vinit A, Stoppin-Mellet V, Devred F, Barbier P, Peyrot V, Sayas CL, Avila J, Peris L, Andrieux A, Serre L, Fourest-Lieuvin A, Arnal I. Tau antagonizes end-binding protein tracking at microtubule ends through a phosphorylation-dependent mechanism. Mol Biol Cell 2016; 27:2924-34. [PMID: 27466319 PMCID: PMC5042579 DOI: 10.1091/mbc.e16-01-0029] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 07/22/2016] [Indexed: 02/05/2023] Open
Abstract
Tau antagonizes tracking of end-binding proteins (EBs) at microtubule ends, a process requiring the C-terminal part of EBs and the microtubule-binding sites of tau. The inhibiting activity of tau on EB properties is regulated by tau phosphorylation. The interplay between EBs and tau proteins results in modulation of microtubule dynamics. Proper regulation of microtubule dynamics is essential for cell functions and involves various microtubule-associated proteins (MAPs). Among them, end-binding proteins (EBs) accumulate at microtubule plus ends, whereas structural MAPs bind along the microtubule lattice. Recent data indicate that the structural MAP tau modulates EB subcellular localization in neurons. However, the molecular determinants of EB/tau interaction remain unknown, as is the effect of this interplay on microtubule dynamics. Here we investigate the mechanisms governing EB/tau interaction in cell-free systems and cellular models. We find that tau inhibits EB tracking at microtubule ends. Tau and EBs form a complex via the C-terminal region of EBs and the microtubule-binding sites of tau. These two domains are required for the inhibitory activity of tau on EB localization to microtubule ends. Moreover, the phosphomimetic mutation S262E within tau microtubule-binding sites impairs EB/tau interaction and prevents the inhibitory effect of tau on EB comets. We further show that microtubule dynamic parameters vary, depending on the combined activities of EBs and tau proteins. Overall our results demonstrate that tau directly antagonizes EB function through a phosphorylation-dependent mechanism. This study highlights a novel role for tau in EB regulation, which might be impaired in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sacnicte Ramirez-Rios
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France
| | - Eric Denarier
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France CEA, BIG-GPC, F-38000 Grenoble France
| | - Eléa Prezel
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France
| | - Angélique Vinit
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France
| | - Virginie Stoppin-Mellet
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France
| | - François Devred
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Faculté de Pharmacie, F-13385 Marseille, France
| | - Pascale Barbier
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Faculté de Pharmacie, F-13385 Marseille, France
| | - Vincent Peyrot
- Aix-Marseille Université, INSERM, CRO2 UMR_S 911, Faculté de Pharmacie, F-13385 Marseille, France
| | - Carmen Laura Sayas
- Centro de Investigaciones Biomédicas de Canarias entro de Investigaciones Biomédicas de Canarias, Instituto de Tecnologías Biomédicas, Universidad de La Laguna, 38071 Tenerife, Spain
| | - Jesus Avila
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), 28029 Madrid, Spain
| | - Leticia Peris
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France
| | - Annie Andrieux
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France CEA, BIG-GPC, F-38000 Grenoble France
| | - Laurence Serre
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France
| | - Anne Fourest-Lieuvin
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France CEA, BIG-GPC, F-38000 Grenoble France
| | - Isabelle Arnal
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, F-38000 Grenoble, France INSERM, U1216, F-38000 Grenoble, France
| |
Collapse
|
27
|
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
|
28
|
Control of microtubule organization and dynamics: two ends in the limelight. Nat Rev Mol Cell Biol 2015; 16:711-26. [PMID: 26562752 DOI: 10.1038/nrm4084] [Citation(s) in RCA: 651] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microtubules have fundamental roles in many essential biological processes, including cell division and intracellular transport. They assemble and disassemble from their two ends, denoted the plus end and the minus end. Significant advances have been made in our understanding of microtubule plus-end-tracking proteins (+TIPs) such as end-binding protein 1 (EB1), XMAP215, selected kinesins and dynein. By contrast, information on microtubule minus-end-targeting proteins (-TIPs), such as the calmodulin-regulated spectrin-associated proteins (CAMSAPs) and Patronin, has only recently started to emerge. Here, we review our current knowledge of factors, including microtubule-targeting agents, that associate with microtubule ends to control the dynamics and function of microtubules during the cell cycle and development.
Collapse
|
29
|
ADNP/ADNP2 expression in oligodendrocytes: implication for myelin-related neurodevelopment. J Mol Neurosci 2015; 57:304-13. [DOI: 10.1007/s12031-015-0640-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
30
|
Hidalgo-de-Quintana J, Schwarz N, Meschede IP, Stern-Schneider G, Powner MB, Morrison EE, Futter CE, Wolfrum U, Cheetham ME, van der Spuy J. The Leber congenital amaurosis protein AIPL1 and EB proteins co-localize at the photoreceptor cilium. PLoS One 2015; 10:e0121440. [PMID: 25799540 PMCID: PMC4370678 DOI: 10.1371/journal.pone.0121440] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 02/12/2015] [Indexed: 11/21/2022] Open
Abstract
Purpose The aim of this study was to investigate the interaction and co-localization of novel interacting proteins with the Leber congenital amaurosis (LCA) associated protein aryl hydrocarbon receptor interacting protein-like 1 (AIPL1). Methods The CytoTrapXR yeast two-hybrid system was used to screen a bovine retinal cDNA library. A novel interaction between AIPL1 and members of the family of EB proteins was confirmed by directed yeast two-hybrid analysis and co-immunoprecipitation assays. The localization of AIPL1 and the EB proteins in cultured cells and in retinal cryosections was examined by immunofluorescence microscopy and cryo-immunogold electron microscopy. Results Yeast two-hybrid (Y2H) analysis identified the interaction between AIPL1 and the EB proteins, EB1 and EB3. EB1 and EB3 were specifically co-immunoprecipitated with AIPL1 from SK-N-SH neuroblastoma cells. In directed 1:1 Y2H analysis, the interaction of EB1 with AIPL1 harbouring the LCA-causing mutations A197P, C239R and W278X was severely compromised. Immunofluorescent confocal microscopy revealed that AIPL1 did not co-localize with endogenous EB1 at the tips of microtubules, endogenous EB1 at the microtubule organising centre following disruption of the microtubule network, or with endogenous β-tubulin. Moreover, AIPL1 did not localize to primary cilia in ARPE-19 cells, whereas EB1 co-localized with the centrosomal marker pericentrin at the base of primary cilia. However, both AIPL1 and the EB proteins, EB1 and EB3, co-localized with centrin-3 in the connecting cilium of photoreceptor cells. Cryo-immunogold electron microscopy confirmed the co-localization of AIPL1 and EB1 in the connecting cilia in human retinal photoreceptors. Conclusions AIPL1 and the EB proteins, EB1 and EB3, localize at the connecting cilia of retinal photoreceptor cells, but do not co-localize in the cellular microtubule network or in primary cilia in non-retinal cells. These findings suggest that AIPL1 function in these cells is not related to the role of EB proteins in microtubule dynamics or primary ciliogenesis, but that their association may be related to a specific role in the specialized cilia apparatus of retinal photoreceptors.
Collapse
Affiliation(s)
- Juan Hidalgo-de-Quintana
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Nele Schwarz
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Ingrid P. Meschede
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Gabriele Stern-Schneider
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Michael B. Powner
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Ewan E. Morrison
- Section of Ophthalmology and Neuroscience, Leeds Institute of Molecular Medicine, St James’s University Hospital, Leeds, United Kingdom
| | - Clare E. Futter
- Department of Cell Biology, UCL Institute of Ophthalmology, London, United Kingdom
| | - Uwe Wolfrum
- Cell and Matrix Biology, Institute of Zoology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Michael E. Cheetham
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
| | - Jacqueline van der Spuy
- Department of Ocular Biology and Therapeutics, UCL Institute of Ophthalmology, London, United Kingdom
- * E-mail:
| |
Collapse
|
31
|
Ferreira JG, Pereira AL, Maiato H. Microtubule plus-end tracking proteins and their roles in cell division. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 309:59-140. [PMID: 24529722 DOI: 10.1016/b978-0-12-800255-1.00002-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microtubules are cellular components that are required for a variety of essential processes such as cell motility, mitosis, and intracellular transport. This is possible because of the inherent dynamic properties of microtubules. Many of these properties are tightly regulated by a number of microtubule plus-end-binding proteins or +TIPs. These proteins recognize the distal end of microtubules and are thus in the right context to control microtubule dynamics. In this review, we address how microtubule dynamics are regulated by different +TIP families, focusing on how functionally diverse +TIPs spatially and temporally regulate microtubule dynamics during animal cell division.
Collapse
Affiliation(s)
- Jorge G Ferreira
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal; Cell Division Unit, Department of Experimental Biology, University of Porto, Porto, Portugal
| | - Ana L Pereira
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| | - Helder Maiato
- Chromosome Instability & Dynamics Laboratory, Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal; Cell Division Unit, Department of Experimental Biology, University of Porto, Porto, Portugal.
| |
Collapse
|
32
|
The NAP motif of activity-dependent neuroprotective protein (ADNP) regulates dendritic spines through microtubule end binding proteins. Mol Psychiatry 2014; 19:1115-24. [PMID: 25178163 DOI: 10.1038/mp.2014.97] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/14/2014] [Accepted: 07/08/2014] [Indexed: 12/29/2022]
Abstract
The NAP motif of activity-dependent neuroprotective protein (ADNP) enhanced memory scores in patients suffering from mild cognitive impairment and protected activities of daily living in schizophrenia patients, while fortifying microtubule (MT)-dependent axonal transport, in mice and flies. The question is how does NAP fortify MTs? Our sequence analysis identified the MT end-binding protein (EB1)-interacting motif SxIP (SIP, Ser-Ile-Pro) in ADNP/NAP and showed specific SxIP binding sites in all members of the EB protein family (EB1-3). Others found that EB1 enhancement of neurite outgrowth is attenuated by EB2, while EB3 interacts with postsynaptic density protein 95 (PSD-95) to modulate dendritic plasticity. Here, NAP increased PSD-95 expression in dendritic spines, which was inhibited by EB3 silencing. EB1 or EB3, but not EB2 silencing inhibited NAP-mediated cell protection, which reflected NAP binding specificity. NAPVSKIPQ (SxIP=SKIP), but not NAPVAAAAQ mimicked NAP activity. ADNP, essential for neuronal differentiation and brain formation in mouse, a member of the SWI/SNF chromatin remodeling complex and a major protein mutated in autism and deregulated in schizophrenia in men, showed similar EB interactions, which were enhanced by NAP treatment. The newly identified shared MT target of NAP/ADNP is directly implicated in synaptic plasticity, explaining the breadth and efficiency of neuroprotective/neurotrophic capacities.
Collapse
|
33
|
Gireesh KK, Sreeja JS, Chakraborti S, Singh P, Thomas GE, Gupta H, Manna T. Microtubule +TIP protein EB1 binds to GTP and undergoes dissociation from dimer to monomer on binding GTP. Biochemistry 2014; 53:5551-7. [PMID: 25111064 DOI: 10.1021/bi5007942] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The +TIP protein EB1 autonomously tracks the growing plus end of microtubules and regulates plus-end dynamics. Previous studies have indicated that EB1 can recognize GTP-bound tubulin structures at the plus end, and it localizes on the microtubule surface at a site close to the exchangeable GTP-binding site of tubulin. Although the GTP-dependent structural change in tubulin has been demonstrated to be a critical determinant for recognition of plus ends by EB1, the effect of GTP on the structure of EB1 has remained unclear. Here, we have used spectroscopic, calorimetric, and biochemical methods to analyze the effect of GTP on EB1 in vitro. Isothermal titration calorimetry and tryptophan fluorescence quenching experiments demonstrated that EB1 binds to GTP with a dissociation constant ~30 μM. Circular dichroism measurements showed that EB1 undergoes changes in its secondary structure on binding GTP. Size-exclusion chromatography and urea-induced unfolding analyses revealed that GTP binding induces dissociation of the EB1 dimer to monomers. Size-exclusion chromatography followed by biochemical analysis further determined that EB1-GTP binding involves association of approximately one molecule of GTP per EB1 monomer. The results reveal a hitherto unknown GTP-dependent mechanism of dimer-to-monomer transition in EB1 and further implicate its possible role in regulating the stability of the EB1 dimer vs monomer as well as plus-end regulation in cells.
Collapse
Affiliation(s)
- K K Gireesh
- School of Biology, Indian Institute of Science Education and Research, Thiruvananthapuram , CET Campus, Thiruvananthapuram, Kerala 695016, India
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Cilia and flagella are surface-exposed, finger-like organelles whose core consists of a microtubule (MT)-based axoneme that grows from a modified centriole, the basal body. Cilia are found on the surface of many eukaryotic cells and play important roles in cell motility and in coordinating a variety of signaling pathways during growth, development, and tissue homeostasis. Defective cilia have been linked to a number of developmental disorders and diseases, collectively called ciliopathies. Cilia are dynamic organelles that assemble and disassemble in tight coordination with the cell cycle. In most cells, cilia are assembled during growth arrest in a multistep process involving interaction of vesicles with appendages present on the distal end of mature centrioles, and addition of tubulin and other building blocks to the distal tip of the basal body and growing axoneme; these building blocks are sorted through a region at the cilium base known as the ciliary necklace, and then transported via intraflagellar transport (IFT) along the axoneme toward the tip for assembly. After assembly, the cilium frequently continues to turn over and incorporate tubulin at its distal end in an IFT-dependent manner. Prior to cell division, the cilia are usually resorbed to liberate centrosomes for mitotic spindle pole formation. Here, we present an overview of the main cytoskeletal structures associated with cilia and centrioles with emphasis on the MT-associated appendages, fibers, and filaments at the cilium base and tip. The composition and possible functions of these structures are discussed in relation to cilia assembly, disassembly, and length regulation.
Collapse
Affiliation(s)
- Lotte B Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
35
|
Sen I, Veprintsev D, Akhmanova A, Steinmetz MO. End binding proteins are obligatory dimers. PLoS One 2013; 8:e74448. [PMID: 24040250 PMCID: PMC3765442 DOI: 10.1371/journal.pone.0074448] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/03/2013] [Indexed: 02/02/2023] Open
Abstract
End binding (EB) proteins are responsible for the recruitment of an array of microtubule plus-end tracking proteins (+TIPs) to growing microtubules ends. EBs encompass an N-terminal calponin homology domain that confers microtubule tip tracking activity to the protein. The C-terminal domain of EBs contains a coiled coil that mediates the parallel dimerization of EB monomers. This part of the protein is also responsible for partner binding. While dimerization is not essential for microtubule tip tracking by EBs it is a prerequisite for +TIP partner binding. The concentration of EBs in cells has been estimated to be in the range of hundreds of nanomoles. In contrast, in in vitro single molecule experiments EB concentrations of subnanomoles are employed. From a mechanistic point of view it is important to assess the oligomerization state of EBs at physiologically and experimentally relevant protein concentrations, in particular if the goal of a study is to model the behavior of EB-dependent dynamic +TIP networks. Here we have determined the stability of the EB1 and EB3 dimers using multi-angle light scattering and fluorescence analytical ultracentrifugation. We show that these EBs form stable dimers and do not dissociate even at very low nanomolar concentrations. The dimers remained stable at both room temperature as well as at the physiologically relevant temperature of 37°C. Together, our results reveal that EBs are obligatory dimers, a conclusion that has implications for the mechanistic understanding of these key proteins involved in the orchestration of dynamic protein networks at growing microtubule ends.
Collapse
Affiliation(s)
- Indrani Sen
- Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen PSI, Switzerland
| | - Dmitry Veprintsev
- Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen PSI, Switzerland
| | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Michel O. Steinmetz
- Laboratory of Biomolecular Research, Paul Scherrer Institut, Villigen PSI, Switzerland
- * E-mail:
| |
Collapse
|
36
|
Larsen J, Grigoriev I, Akhmanova A, Pedersen LB. Analysis of microtubule plus-end-tracking proteins in cilia. Methods Enzymol 2013; 524:105-22. [PMID: 23498737 DOI: 10.1016/b978-0-12-397945-2.00007-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The microtubule (MT) plus-end-tracking proteins (+TIPs) belonging to the end binding (EB) protein family have been studied extensively in the context of cytoplasmic MTs and were shown to play pivotal roles in regulating MT dynamics and in recruiting other +TIPs to MT ends. Early studies in the green alga Chlamydomonas reinhardtii showed that EB1 localizes to the distal flagellum tip and the basal body, and subsequent studies using green fluorescent protein-tagged fusion proteins have demonstrated similar localization of EBs in other ciliated organisms, including mammalian cells as demonstrated here. Functional analysis of EBs in cultured mammalian cells revealed that EB1 and EB3 are required for biogenesis of primary cilia. Although mammalian EB3 localizes to the tip of some cilia and induces cilium elongation, EBs primarily seem to promote ciliogenesis via MT minus-end anchoring at the basal body, in turn facilitating vesicular trafficking to the cilium base. Moreover, mammalian EBs were shown to interact with several proteins implicated in MT minus-end anchoring and/or vesicular trafficking to cilia. Recent work suggests that apart from EBs, additional +TIPs may be present at the distal tip of cilia where they could regulate axoneme assembly, stability, or disassembly.
Collapse
Affiliation(s)
- Jesper Larsen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|
37
|
Ferreira JG, Pereira AJ, Akhmanova A, Maiato H. Aurora B spatially regulates EB3 phosphorylation to coordinate daughter cell adhesion with cytokinesis. ACTA ACUST UNITED AC 2013; 201:709-24. [PMID: 23712260 PMCID: PMC3664705 DOI: 10.1083/jcb.201301131] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
During mitosis, human cells round up, decreasing their adhesion to extracellular substrates. This must be quickly reestablished by poorly understood cytoskeleton remodeling mechanisms that prevent detachment from epithelia, while ensuring the successful completion of cytokinesis. Here we show that the microtubule end-binding (EB) proteins EB1 and EB3 play temporally distinct roles throughout cell division. Whereas EB1 was involved in spindle orientation before anaphase, EB3 was required for stabilization of focal adhesions and coordinated daughter cell spreading during mitotic exit. Additionally, EB3 promoted midbody microtubule stability and, consequently, midbody stabilization necessary for efficient cytokinesis. Importantly, daughter cell adhesion and cytokinesis completion were spatially regulated by distinct states of EB3 phosphorylation on serine 176 by Aurora B. This EB3 phosphorylation was enriched at the midbody and shown to control cortical microtubule growth. These findings uncover differential roles of EB proteins and explain the importance of an Aurora B phosphorylation gradient for the spatiotemporal regulation of microtubule function during mitotic exit and cytokinesis.
Collapse
Affiliation(s)
- Jorge G Ferreira
- Chromosome Instability and Dynamics Laboratory, Instituto de Biologia Molecular e Celular (IBMC), University of Porto, Porto, Portugal
| | | | | | | |
Collapse
|
38
|
Stenner F, Liewen H, Göttig S, Henschler R, Markuly N, Kleber S, Faust M, Mischo A, Bauer S, Zweifel M, Knuth A, Renner C, Wadle A. RP1 is a phosphorylation target of CK2 and is involved in cell adhesion. PLoS One 2013; 8:e67595. [PMID: 23844040 PMCID: PMC3701075 DOI: 10.1371/journal.pone.0067595] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 05/22/2013] [Indexed: 12/24/2022] Open
Abstract
RP1 (synonym: MAPRE2, EB2) is a member of the microtubule binding EB1 protein family, which interacts with APC, a key regulatory molecule in the Wnt signalling pathway. While the other EB1 proteins are well characterized the cellular function and regulation of RP1 remain speculative to date. However, recently RP1 has been implicated in pancreatic cancerogenesis. CK2 is a pleiotropic kinase involved in adhesion, proliferation and anti-apoptosis. Overexpression of protein kinase CK2 is a hallmark of many cancers and supports the malignant phenotype of tumor cells. In this study we investigate the interaction of protein kinase CK2 with RP1 and demonstrate that CK2 phosphorylates RP1 at Ser236 in vitro. Stable RP1 expression in cell lines leads to a significant cleavage and down-regulation of N-cadherin and impaired adhesion. Cells expressing a Phospho-mimicking point mutant RP1-ASP236 show a marked decrease of adhesion to endothelial cells under shear stress. Inversely, we found that the cells under shear stress downregulate endogenous RP1, most likely to improve cellular adhesion. Accordingly, when RP1 expression is suppressed by shRNA, cells lacking RP1 display significantly increased cell adherence to surfaces. In summary, RP1 phosphorylation at Ser236 by CK2 seems to play a significant role in cell adhesion and might initiate new insights in the CK2 and EB1 family protein association.
Collapse
Affiliation(s)
- Frank Stenner
- Division of Oncology, University Hospital Zurich, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Alberico EO, Lyons DF, Murphy RJ, Philip JT, Duan AR, Correia JJ, Goodson HV. Biochemical evidence that human EB1 does not bind preferentially to the microtubule seam. Cytoskeleton (Hoboken) 2013; 70:317-27. [PMID: 23864329 DOI: 10.1002/cm.21108] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 03/14/2013] [Accepted: 03/15/2013] [Indexed: 11/08/2022]
Abstract
EB1 is a highly conserved microtubule (MT) plus end tracking protein (+TIP) involved in regulating MT dynamics, but the mechanisms of its effects on MT polymerization remain undefined. Resolving this question requires understanding how EB1 interacts with MTs. Previous electron microscopy of the S. pombe EB1 homolog Mal3p suggested that Mal3p binds specifically to the MT seam, implying that EB1 family members promote MT polymerization by stabilizing the seam. However, more recent electron microscopy indicates that Mal3p binds everywhere except the seam. Neither set of experiments investigated the behavior of human EB1, or provided an explanation for why these studies arrived at different answers. To resolve these questions, we have used a combination of MT-binding assays and theoretical modeling with MTBindingSim. Our results indicate that human EB1 binds to the lattice, consistent with the recent Mal3p results, and show that Mal3p-binding assays that were previously interpreted as evidence for preferential seam binding are equally consistent with weak lattice binding. In addition, we used analytical ultracentrifugation to investigate the possibility that the EB1 monomer-dimer equilibrium might contribute to EB1 binding behavior, and determined that the EB1 dimerization dissociation constant is approximately 90 nM. We and others find that the cellular concentration of EB1 is on the order of 200 nM, suggesting that a portion of EB1 may be monomeric at physiological concentrations. These observations lead us to suggest that regulation of EB1 dimerization might play a role in controlling EB1 function.
Collapse
Affiliation(s)
- Emily O Alberico
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Komarova YA, Huang F, Geyer M, Daneshjou N, Garcia A, Idalino L, Kreutz B, Mehta D, Malik AB. VE-cadherin signaling induces EB3 phosphorylation to suppress microtubule growth and assemble adherens junctions. Mol Cell 2012; 48:914-25. [PMID: 23159740 DOI: 10.1016/j.molcel.2012.10.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 07/23/2012] [Accepted: 10/09/2012] [Indexed: 11/28/2022]
Abstract
Vascular endothelial (VE)-cadherin homophilic adhesion controls endothelial barrier permeability through assembly of adherens junctions (AJs). We observed that loss of VE-cadherin-mediated adhesion induced the activation of Src and phospholipase C (PLC)γ2, which mediated Ca(2+) release from endoplasmic reticulum (ER) stores, resulting in activation of calcineurin (CaN), a Ca(2+)-dependent phosphatase. Downregulation of CaN activity induced phosphorylation of serine 162 in end binding (EB) protein 3. This phospho-switch was required to destabilize the EB3 dimer, suppress microtubule (MT) growth, and assemble AJs. The phospho-defective S162A EB3 mutant, in contrast, induced MT growth in confluent endothelial monolayers and disassembled AJs. Thus, VE-cadherin outside-in signaling regulates cytosolic Ca(2+) homeostasis and EB3 phosphorylation, which are required for assembly of AJs. These results identify a pivotal function of VE-cadherin homophilic interaction in modulating endothelial barrier through the tuning of MT dynamics.
Collapse
Affiliation(s)
- Yulia A Komarova
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Welburn JPI, Cheeseman IM. The microtubule-binding protein Cep170 promotes the targeting of the kinesin-13 depolymerase Kif2b to the mitotic spindle. Mol Biol Cell 2012; 23:4786-95. [PMID: 23087211 PMCID: PMC3521686 DOI: 10.1091/mbc.e12-03-0214] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The N-terminus of the kinesin-13 family (Kif2a, Kif2b, Kif2c) is the primary localization determinant. However, the C-terminus of Kif2b associates with Cep170 and Cep170R to create targeting specificity. Cep170 has microtubule-binding properties in vitro and provides a second microtubule-binding site to Kif2b to target it to the spindle. Microtubule dynamics are essential throughout mitosis to ensure correct chromosome segregation. Microtubule depolymerization is controlled in part by microtubule depolymerases, including the kinesin-13 family of proteins. In humans, there are three closely related kinesin-13 isoforms (Kif2a, Kif2b, and Kif2c/MCAK), which are highly conserved in their primary sequences but display distinct localization and nonoverlapping functions. Here we demonstrate that the N-terminus is a primary determinant of kinesin-13 localization. However, we also find that differences in the C-terminus alter the properties of kinesin-13, in part by facilitating unique protein–protein interactions. We identify the spindle-localized proteins Cep170 and Cep170R (KIAA0284) as specifically associating with Kif2b. Cep170 binds to microtubules in vitro and provides Kif2b with a second microtubule-binding site to target it to the spindle. Thus the intrinsic properties of kinesin-13s and extrinsic factors such as their associated proteins result in the diversity and specificity within the kinesin-13 depolymerase family.
Collapse
Affiliation(s)
- Julie P I Welburn
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| | | |
Collapse
|
42
|
Kumar P, Wittmann T. +TIPs: SxIPping along microtubule ends. Trends Cell Biol 2012; 22:418-28. [PMID: 22748381 DOI: 10.1016/j.tcb.2012.05.005] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 05/24/2012] [Accepted: 05/25/2012] [Indexed: 01/08/2023]
Abstract
+TIPs are a heterogeneous class of proteins that specifically bind to growing microtubule ends. Because dynamic microtubules are essential for many intracellular processes, +TIPs play important roles in regulating microtubule dynamics and microtubule interactions with other intracellular structures. End-binding proteins (EBs) recognize a structural cap at growing microtubule ends, and have emerged as central adaptors that mediate microtubule plus-end tracking of potentially all other +TIPs. The majority of these +TIPs bind to EBs through a short hydrophobic (S/T)x(I/L)P sequence motif (SxIP) and surrounding electrostatic interactions. These recent discoveries have resulted in a rapid expansion of the number of possible +TIPs. In this review, we outline our current understanding of the molecular mechanism of plus-end tracking and provide an overview of SxIP-recruited +TIPs.
Collapse
Affiliation(s)
- Praveen Kumar
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | | |
Collapse
|
43
|
Buey RM, Sen I, Kortt O, Mohan R, Gfeller D, Veprintsev D, Kretzschmar I, Scheuermann J, Neri D, Zoete V, Michielin O, de Pereda JM, Akhmanova A, Volkmer R, Steinmetz MO. Sequence determinants of a microtubule tip localization signal (MtLS). J Biol Chem 2012; 287:28227-42. [PMID: 22696216 DOI: 10.1074/jbc.m112.373928] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microtubule plus-end-tracking proteins (+TIPs) specifically localize to the growing plus-ends of microtubules to regulate microtubule dynamics and functions. A large group of +TIPs contain a short linear motif, SXIP, which is essential for them to bind to end-binding proteins (EBs) and target microtubule ends. The SXIP sequence site thus acts as a widespread microtubule tip localization signal (MtLS). Here we have analyzed the sequence-function relationship of a canonical MtLS. Using synthetic peptide arrays on membrane supports, we identified the residue preferences at each amino acid position of the SXIP motif and its surrounding sequence with respect to EB binding. We further developed an assay based on fluorescence polarization to assess the mechanism of the EB-SXIP interaction and to correlate EB binding and microtubule tip tracking of MtLS sequences from different +TIPs. Finally, we investigated the role of phosphorylation in regulating the EB-SXIP interaction. Together, our results define the sequence determinants of a canonical MtLS and provide the experimental data for bioinformatics approaches to carry out genome-wide predictions of novel +TIPs in multiple organisms.
Collapse
Affiliation(s)
- Rubén M Buey
- Laboratory of Biomolecular Research, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Schrøder JM, Larsen J, Komarova Y, Akhmanova A, Thorsteinsson RI, Grigoriev I, Manguso R, Christensen ST, Pedersen SF, Geimer S, Pedersen LB. EB1 and EB3 promote cilia biogenesis by several centrosome-related mechanisms. J Cell Sci 2011; 124:2539-51. [PMID: 21768326 DOI: 10.1242/jcs.085852] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The microtubule (MT) plus-end-tracking protein EB1 is required for assembly of primary cilia in mouse fibroblasts, but the mechanisms involved and the roles of the related proteins EB2 and EB3 in ciliogenesis are unknown. Using protein depletion experiments and expression of dominant-negative constructs we show here that EB1 and EB3, but not EB2, are required for assembly of primary cilia in cultured cells. Electron microscopy and live imaging showed that cells lacking EB1 or EB3 are defective in MT minus-end anchoring at the centrosome and/or basal body, and possess abnormally short cilia stumps surrounded by vesicles. Further, GST pull-down assays, mass spectrometry and immunoprecipitation indicated that EB1 and EB3 interact with proteins implicated in MT minus-end anchoring or vesicular trafficking to the cilia base, suggesting that EB1 and EB3 promote ciliogenesis by facilitating such trafficking. In addition, we show that EB3 is localized to the tip of motile cilia in bronchial epithelial cells and affects the formation of centriole-associated rootlet filaments. Collectively, our findings indicate that EBs affect biogenesis of cilia by several centrosome-related mechanisms and support the idea that different EB1-EB3 dimer species have distinct functions within cells.
Collapse
Affiliation(s)
- Jacob M Schrøder
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bjelić S, De Groot CO, Schärer MA, Jaussi R, Bargsten K, Salzmann M, Frey D, Capitani G, Kammerer RA, Steinmetz MO. Interaction of mammalian end binding proteins with CAP-Gly domains of CLIP-170 and p150(glued). J Struct Biol 2011; 177:160-7. [PMID: 22119847 DOI: 10.1016/j.jsb.2011.11.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Revised: 11/07/2011] [Accepted: 11/07/2011] [Indexed: 10/15/2022]
Abstract
End binding proteins (EBs) track growing microtubule ends and play a master role in organizing dynamic protein networks. Mammalian cells express up to three different EBs (EB1, EB2, and EB3). Besides forming homodimers, EB1 and EB3 also assemble into heterodimers. One group of EB-binding partners encompasses proteins that harbor CAP-Gly domains. The binding properties of the different EBs towards CAP-Gly proteins have not been systematically investigated. This information is, however, important to compare and contrast functional differences. Here we analyzed the interactions between CLIP-170 and p150(glued) CAP-Gly domains with the three EB homodimers and the EB1-EB3 heterodimer. Using isothermal titration calorimetry we observed that some EBs bind to the individual CAP-Gly domains with similar affinities while others interact with their targets with pronounced differences. We further found that the two types of CAP-Gly domains use alternative mechanisms to target the C-terminal domains of EBs. We succeeded to solve the crystal structure of a complex composed of a heterodimer of EB1 and EB3 C-termini together with the CAP-Gly domain of p150(glued). Together, our results provide mechanistic insights into the interaction properties of EBs and offer a molecular framework for the systematic investigation of their functional differences in cells.
Collapse
Affiliation(s)
- Saša Bjelić
- Biomolecular Research, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Buey RM, Mohan R, Leslie K, Walzthoeni T, Missimer JH, Menzel A, Bjelić S, Bargsten K, Grigoriev I, Smal I, Meijering E, Aebersold R, Akhmanova A, Steinmetz MO. Insights into EB1 structure and the role of its C-terminal domain for discriminating microtubule tips from the lattice. Mol Biol Cell 2011; 22:2912-23. [PMID: 21737692 PMCID: PMC3154886 DOI: 10.1091/mbc.e11-01-0017] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Revised: 05/12/2011] [Accepted: 06/16/2011] [Indexed: 12/31/2022] Open
Abstract
End-binding proteins (EBs) comprise a conserved family of microtubule plus end-tracking proteins. The concerted action of calponin homology (CH), linker, and C-terminal domains of EBs is important for their autonomous microtubule tip tracking, regulation of microtubule dynamics, and recruitment of numerous partners to microtubule ends. Here we report the detailed structural and biochemical analysis of mammalian EBs. Small-angle X-ray scattering, electron microscopy, and chemical cross-linking in combination with mass spectrometry indicate that EBs are elongated molecules with two interacting CH domains, an arrangement reminiscent of that seen in other microtubule- and actin-binding proteins. Removal of the negatively charged C-terminal tail did not affect the overall conformation of EBs; however, it increased the dwell times of EBs on the microtubule lattice in microtubule tip-tracking reconstitution experiments. An even more stable association with the microtubule lattice was observed when the entire negatively charged C-terminal domain of EBs was replaced by a neutral coiled-coil motif. In contrast, the interaction of EBs with growing microtubule tips was not significantly affected by these C-terminal domain mutations. Our data indicate that long-range electrostatic repulsive interactions between the C-terminus and the microtubule lattice drive the specificity of EBs for growing microtubule ends.
Collapse
Affiliation(s)
- Rubén M. Buey
- Biomolecular Research, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland
| | - Renu Mohan
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CA, Netherlands
| | - Kris Leslie
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CA, Netherlands
| | - Thomas Walzthoeni
- Institute of Molecular Systems Biology, ETH Zürich, 8093 Zürich, Switzerland
- Ph.D. Program in Molecular Life Sciences, University of Zürich/ETH Zürich, 8093 Zürich, Switzerland
| | - John H. Missimer
- Biomolecular Research, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland
| | - Andreas Menzel
- Synchrotron Radiation and Nanotechnology, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland
| | - Saša Bjelić
- Biomolecular Research, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland
| | - Katja Bargsten
- Biomolecular Research, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland
| | - Ilya Grigoriev
- Department of Cell Biology, Erasmus Medical Center, Rotterdam 3000 CA, Netherlands
| | - Ihor Smal
- Biomedical Imaging Group Rotterdam, Department of Medical Informatics and Radiology, Erasmus Medical Center, Rotterdam 3000 CA, Netherlands
| | - Erik Meijering
- Biomedical Imaging Group Rotterdam, Department of Medical Informatics and Radiology, Erasmus Medical Center, Rotterdam 3000 CA, Netherlands
| | - Ruedi Aebersold
- Institute of Molecular Systems Biology, ETH Zürich, 8093 Zürich, Switzerland
- Faculty of Science, University of Zürich, 8093 Zürich, Switzerland
- Competence Center for Systems Physiology and Metabolic Diseases, 8093 Zürich, Switzerland
| | - Anna Akhmanova
- Ph.D. Program in Molecular Life Sciences, University of Zürich/ETH Zürich, 8093 Zürich, Switzerland
| | - Michel O. Steinmetz
- Biomolecular Research, Paul Scherrer Institut, 5232 Villigen PSI, Switzerland
| |
Collapse
|
47
|
Sweet ES, Previtera ML, Fernández JR, Charych EI, Tseng CY, Kwon M, Starovoytov V, Zheng JQ, Firestein BL. PSD-95 alters microtubule dynamics via an association with EB3. J Neurosci 2011; 31:1038-47. [PMID: 21248129 PMCID: PMC3138189 DOI: 10.1523/jneurosci.1205-10.2011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 11/01/2010] [Accepted: 11/09/2010] [Indexed: 11/21/2022] Open
Abstract
Little is known about how the neuronal cytoskeleton is regulated when a dendrite decides whether to branch or not. Previously, we reported that postsynaptic density protein 95 (PSD-95) acts as a stop signal for dendrite branching. It is yet to be elucidated how PSD-95 affects the cytoskeleton and how this regulation relates to the dendritic arbor. Here, we show that the SH3 (src homology 3) domain of PSD-95 interacts with a proline-rich region within the microtubule end-binding protein EB3. Overexpression of PSD-95 or mutant EB3 results in a decreased lifetime of EB3 comets in dendrites. In line with these data, transfected rat neurons show that overexpression of PSD-95 results in less organized microtubules at dendritic branch points and decreased dendritogensis. The interaction between PSD-95 and EB3 elucidates a function for a novel region of EB3 and provides a new and important mechanism for the regulation of microtubules in determining dendritic morphology.
Collapse
Affiliation(s)
- Eric S. Sweet
- Departments of Cell Biology and Neuroscience
- Graduate Program in Neuroscience, Rutgers University, Piscataway, New Jersey 08854-8082, and
| | - Michelle L. Previtera
- Departments of Cell Biology and Neuroscience
- Molecular Biosciences Graduate Program, and
| | - Jose R. Fernández
- Departments of Cell Biology and Neuroscience
- Molecular Biosciences Graduate Program, and
| | | | - Chia-Yi Tseng
- Departments of Cell Biology and Neuroscience
- Graduate Program in Neuroscience, Rutgers University, Piscataway, New Jersey 08854-8082, and
| | - Munjin Kwon
- Departments of Cell Biology and Neuroscience
- Molecular Biosciences Graduate Program, and
| | | | - James Q. Zheng
- Neuroscience and Cell Biology, University of Medicine and Denstistry of New Jersey, Piscataway, New Jersey 08854-5635
| | | |
Collapse
|
48
|
Galjart N. Plus-end-tracking proteins and their interactions at microtubule ends. Curr Biol 2010; 20:R528-37. [PMID: 20620909 DOI: 10.1016/j.cub.2010.05.022] [Citation(s) in RCA: 173] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Microtubules are cytoskeletal elements that are essential for a large number of intracellular processes, including mitosis, cell differentiation and migration, and vesicle transport. In many cells, the microtubule network is organized in a radial manner, with one end of a microtubule (the minus end) embedded near the nucleus and the other end (the plus end) exploring cytoplasmic space, switching between episodes of growth and shrinkage. Mammalian plus-end-tracking proteins (+TIPs) localize to the ends of growing microtubules and regulate both the dynamic behavior of microtubules as well as the interactions of microtubules with other cellular components. Because of these crucial roles, +TIPs and the mechanisms underlying their association with microtubule ends have been intensively investigated. Results indicate that +TIPs reach microtubule ends by motor-mediated transport or diffusion. Individual +TIP molecules exchange rapidly on microtubule end-binding sites that are formed during microtubule polymerization and that have a slower turnover. Most +TIPs associate with the end-binding (EB) proteins, and appear to require these 'core' +TIPs for localization at microtubule ends. Accumulation of +TIPs may also involve structural features of the microtubule end and interactions with other +TIPs. This complexity makes it difficult to assign discrete roles to specific +TIPs. Given that +TIPs concentrate at microtubule ends and that each +TIP binds in a conformationally distinct manner, I propose that the ends of growing microtubules are 'nano-platforms' for productive interactions between selected proteins and that these interactions might persist and be functional elsewhere in the cytoplasm than at the microtubule end at which they originated.
Collapse
Affiliation(s)
- Niels Galjart
- Department of Cell Biology and Genetics, Erasmus MC, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
49
|
Gouveia SM, Akhmanova A. Cell and Molecular Biology of Microtubule Plus End Tracking Proteins. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 285:1-74. [DOI: 10.1016/b978-0-12-381047-2.00001-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|