201
|
Roostalu J, Surrey T. Microtubule nucleation: beyond the template. Nat Rev Mol Cell Biol 2017; 18:702-710. [PMID: 28831203 DOI: 10.1038/nrm.2017.75] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Microtubules are cytoskeletal filaments central to a wide range of essential cellular functions in eukaryotic cells. Consequently, cells need to exert tight control over when, where and how many microtubules are being made. Whereas the regulation of microtubule dynamics is well studied, the molecular mechanisms of microtubule nucleation are still poorly understood. Next to the established master template of nucleation, the γ-tubulin ring complex, other microtubule-associated proteins that affect microtubule dynamic properties have recently been found to contribute to nucleation. It has begun to emerge that the nucleation efficiency is controlled not only by template activity but also by, either additionally or alternatively, the stabilization of the nascent microtubule 'nucleus'. This suggests a simple conceptual framework for the mechanisms regulating microtubule nucleation in cells.
Collapse
Affiliation(s)
| | - Thomas Surrey
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
202
|
Viswanath S, Bonomi M, Kim SJ, Klenchin VA, Taylor KC, Yabut KC, Umbreit NT, Van Epps HA, Meehl J, Jones MH, Russel D, Velazquez-Muriel JA, Winey M, Rayment I, Davis TN, Sali A, Muller EG. The molecular architecture of the yeast spindle pole body core determined by Bayesian integrative modeling. Mol Biol Cell 2017; 28:3298-3314. [PMID: 28814505 PMCID: PMC5687031 DOI: 10.1091/mbc.e17-06-0397] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/07/2017] [Accepted: 08/09/2017] [Indexed: 12/31/2022] Open
Abstract
A model of the core of the yeast spindle pole body (SPB) was created by a Bayesian modeling approach that integrated a diverse data set of biophysical, biochemical, and genetic information. The model led to a proposed pathway for the assembly of Spc110, a protein related to pericentrin, and a mechanism for how calmodulin strengthens the SPB during mitosis. Microtubule-organizing centers (MTOCs) form, anchor, and stabilize the polarized network of microtubules in a cell. The central MTOC is the centrosome that duplicates during the cell cycle and assembles a bipolar spindle during mitosis to capture and segregate sister chromatids. Yet, despite their importance in cell biology, the physical structure of MTOCs is poorly understood. Here we determine the molecular architecture of the core of the yeast spindle pole body (SPB) by Bayesian integrative structure modeling based on in vivo fluorescence resonance energy transfer (FRET), small-angle x-ray scattering (SAXS), x-ray crystallography, electron microscopy, and two-hybrid analysis. The model is validated by several methods that include a genetic analysis of the conserved PACT domain that recruits Spc110, a protein related to pericentrin, to the SPB. The model suggests that calmodulin can act as a protein cross-linker and Spc29 is an extended, flexible protein. The model led to the identification of a single, essential heptad in the coiled-coil of Spc110 and a minimal PACT domain. It also led to a proposed pathway for the integration of Spc110 into the SPB.
Collapse
Affiliation(s)
- Shruthi Viswanath
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158
| | - Massimiliano Bonomi
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158 .,Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Seung Joong Kim
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158
| | - Vadim A Klenchin
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Keenan C Taylor
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - King C Yabut
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Neil T Umbreit
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | | | - Janet Meehl
- Department of Molecular, Cellular and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309
| | - Michele H Jones
- Department of Molecular, Cellular and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309
| | - Daniel Russel
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158
| | - Javier A Velazquez-Muriel
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158
| | - Mark Winey
- Department of Molecular, Cellular and Developmental Biology, University of Colorado-Boulder, Boulder, CO 80309
| | - Ivan Rayment
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706
| | - Trisha N Davis
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158
| | - Eric G Muller
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| |
Collapse
|
203
|
Roberts B, Haupt A, Tucker A, Grancharova T, Arakaki J, Fuqua MA, Nelson A, Hookway C, Ludmann SA, Mueller IA, Yang R, Horwitz R, Rafelski SM, Gunawardane RN. Systematic gene tagging using CRISPR/Cas9 in human stem cells to illuminate cell organization. Mol Biol Cell 2017; 28:2854-2874. [PMID: 28814507 PMCID: PMC5638588 DOI: 10.1091/mbc.e17-03-0209] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
The generation of a collection of human induced pluripotent stem cell (hiPSC) lines expressing endogenously GFP-tagged proteins using CRISPR/Cas9 methods is described. The methods used and the genomic and cell biological data validating the GFP-tagged hiPSC lines are also presented. We present a CRISPR/Cas9 genome-editing strategy to systematically tag endogenous proteins with fluorescent tags in human induced pluripotent stem cells (hiPSC). To date, we have generated multiple hiPSC lines with monoallelic green fluorescent protein tags labeling 10 proteins representing major cellular structures. The tagged proteins include alpha tubulin, beta actin, desmoplakin, fibrillarin, nuclear lamin B1, nonmuscle myosin heavy chain IIB, paxillin, Sec61 beta, tight junction protein ZO1, and Tom20. Our genome-editing methodology using Cas9/crRNA ribonuclear protein and donor plasmid coelectroporation, followed by fluorescence-based enrichment of edited cells, typically resulted in <0.1–4% homology-directed repair (HDR). Twenty-five percent of clones generated from each edited population were precisely edited. Furthermore, 92% (36/39) of expanded clonal lines displayed robust morphology, genomic stability, expression and localization of the tagged protein to the appropriate subcellular structure, pluripotency-marker expression, and multilineage differentiation. It is our conclusion that, if cell lines are confirmed to harbor an appropriate gene edit, pluripotency, differentiation potential, and genomic stability are typically maintained during the clonal line–generation process. The data described here reveal general trends that emerged from this systematic gene-tagging approach. Final clonal lines corresponding to each of the 10 cellular structures are now available to the research community.
Collapse
Affiliation(s)
| | - Amanda Haupt
- Allen Institute for Cell Science, Seattle, WA 98109
| | | | | | - Joy Arakaki
- Allen Institute for Cell Science, Seattle, WA 98109
| | | | | | | | | | | | - Ruian Yang
- Allen Institute for Cell Science, Seattle, WA 98109
| | - Rick Horwitz
- Allen Institute for Cell Science, Seattle, WA 98109
| | | | | |
Collapse
|
204
|
Takatani S, Ozawa S, Yagi N, Hotta T, Hashimoto T, Takahashi Y, Takahashi T, Motose H. Directional cell expansion requires NIMA-related kinase 6 (NEK6)-mediated cortical microtubule destabilization. Sci Rep 2017; 7:7826. [PMID: 28798328 PMCID: PMC5552743 DOI: 10.1038/s41598-017-08453-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/12/2017] [Indexed: 11/09/2022] Open
Abstract
Plant cortical microtubules align perpendicular to the growth axis to determine the direction of cell growth. However, it remains unclear how plant cells form well-organized cortical microtubule arrays in the absence of a centrosome. In this study, we investigated the functions of Arabidopsis NIMA-related kinase 6 (NEK6), which regulates microtubule organization during anisotropic cell expansion. Quantitative analysis of hypocotyl cell growth in the nek6-1 mutant demonstrated that NEK6 suppresses ectopic outgrowth and promotes cell elongation in different regions of the hypocotyl. Loss of NEK6 function led to excessive microtubule waving and distortion, implying that NEK6 suppresses the aberrant cortical microtubules. Live cell imaging showed that NEK6 localizes to the microtubule lattice and to the shrinking plus and minus ends of microtubules. In agreement with this observation, the induced overexpression of NEK6 reduced and disorganized cortical microtubules and suppressed cell elongation. Furthermore, we identified five phosphorylation sites in β-tubulin that serve as substrates for NEK6 in vitro. Alanine substitution of the phosphorylation site Thr166 promoted incorporation of mutant β-tubulin into microtubules. Taken together, these results suggest that NEK6 promotes directional cell growth through phosphorylation of β-tubulin and the resulting destabilization of cortical microtubules.
Collapse
Affiliation(s)
- Shogo Takatani
- Department of Biological Science, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan
| | - Shinichiro Ozawa
- Department of Biological Science, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan.,Japan Science and Technology Agency, 4-1-8 Kawaguchi, Saitama, 332-0012, Japan
| | - Noriyoshi Yagi
- Graduate School of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan.,Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Takashi Hotta
- Graduate School of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan.,Department of Embryology, Carnegie Institution for Science, 3520 San Martin Drive, Baltimore, MD, 21218, USA
| | - Takashi Hashimoto
- Graduate School of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Yuichiro Takahashi
- Department of Biological Science, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan.,Japan Science and Technology Agency, 4-1-8 Kawaguchi, Saitama, 332-0012, Japan
| | - Taku Takahashi
- Department of Biological Science, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan
| | - Hiroyasu Motose
- Department of Biological Science, Graduate School of Natural Science and Technology, Okayama University, 3-1-1 Tsushimanaka, Okayama, 700-8530, Japan.
| |
Collapse
|
205
|
Vleugel M, Kok M, Dogterom M. Understanding force-generating microtubule systems through in vitro reconstitution. Cell Adh Migr 2017; 10:475-494. [PMID: 27715396 PMCID: PMC5079405 DOI: 10.1080/19336918.2016.1241923] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Microtubules switch between growing and shrinking states, a feature known as dynamic instability. The biochemical parameters underlying dynamic instability are modulated by a wide variety of microtubule-associated proteins that enable the strict control of microtubule dynamics in cells. The forces generated by controlled growth and shrinkage of microtubules drive a large range of processes, including organelle positioning, mitotic spindle assembly, and chromosome segregation. In the past decade, our understanding of microtubule dynamics and microtubule force generation has progressed significantly. Here, we review the microtubule-intrinsic process of dynamic instability, the effect of external factors on this process, and how the resulting forces act on various biological systems. Recently, reconstitution-based approaches have strongly benefited from extensive biochemical and biophysical characterization of individual components that are involved in regulating or transmitting microtubule-driven forces. We will focus on the current state of reconstituting increasingly complex biological systems and provide new directions for future developments.
Collapse
Affiliation(s)
- Mathijs Vleugel
- a Department of Bionanoscience , Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft Institute of Technology , Delft , The Netherlands
| | - Maurits Kok
- a Department of Bionanoscience , Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft Institute of Technology , Delft , The Netherlands
| | - Marileen Dogterom
- a Department of Bionanoscience , Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft Institute of Technology , Delft , The Netherlands
| |
Collapse
|
206
|
Abstract
Microtubules (MTs) form a rapidly adaptable network of filaments that radiate throughout the cell. These dynamic arrays facilitate a wide range of cellular processes, including the capture, transport, and spatial organization of cargos and organelles, as well as changes in cell shape, polarity, and motility. Nucleating from MT-organizing centers, including but by no means limited to the centrosome, MTs undergo rapid transitions through phases of growth, pause, and catastrophe, continuously exploring and adapting to the intracellular environment. Subsets of MTs can become stabilized in response to environmental cues, acquiring distinguishing posttranslational modifications and performing discrete functions as specialized tracks for cargo trafficking. The dynamic behavior and organization of the MT array is regulated by MT-associated proteins (MAPs), which include a subset of highly specialized plus-end-tracking proteins (+TIPs) that respond to signaling cues to alter MT behavior. As pathogenic cargos, viruses require MTs to transport to and from their intracellular sites of replication. While interactions with and functions for MT motor proteins are well characterized and extensively reviewed for many viruses, this review focuses on MT filaments themselves. Changes in the spatial organization and dynamics of the MT array, mediated by virus- or host-induced changes to MT regulatory proteins, not only play a central role in the intracellular transport of virus particles but also regulate a wider range of processes critical to the outcome of infection.
Collapse
|
207
|
Chen JV, Buchwalter RA, Kao LR, Megraw TL. A Splice Variant of Centrosomin Converts Mitochondria to Microtubule-Organizing Centers. Curr Biol 2017; 27:1928-1940.e6. [PMID: 28669756 DOI: 10.1016/j.cub.2017.05.090] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 05/02/2017] [Accepted: 05/31/2017] [Indexed: 11/25/2022]
Abstract
Non-centrosomal microtubule organizing centers (MTOCs) direct microtubule (MT) organization to exert diverse cell-type-specific functions. In Drosophila spermatids, the giant mitochondria provide structural platforms for MT reorganization to support elongation of the extremely long sperm. However, the molecular basis for this mitochondrial MTOC and other non-centrosomal MTOCs has not been discerned. Here we report that Drosophila centrosomin (cnn) expresses two major protein variants: the centrosomal form (CnnC) and a non-centrosomal form in testes (CnnT). CnnC is established as essential for functional centrosomes, the major MTOCs in animal cells. We show that CnnT is expressed exclusively in testes by alternative splicing and localizes to giant mitochondria in spermatids. In cell culture, CnnT targets to the mitochondrial surface, recruits the MT nucleator γ-tubulin ring complex (γ-TuRC), and is sufficient to convert mitochondria to MTOCs independent of core pericentriolar proteins that regulate MT assembly at centrosomes. We mapped two separate domains in CnnT: one that is necessary and sufficient to target it to mitochondria and another that is necessary and sufficient to recruit γ-TuRCs and nucleate MTs. In elongating spermatids, CnnT forms speckles on the giant mitochondria that are required to recruit γ-TuRCs to organize MTs and support spermiogenesis. This molecular characterization of the mitochondrial MTOC defines a minimal molecular requirement for MTOC generation and implicates the potent role of Cnn (or its related) proteins in the direct regulation of MT assembly and organization of non-centrosomal MTOCs.
Collapse
Affiliation(s)
- Jieyan V Chen
- Department of Biomedical Sciences, Florida State University, 1115 West Call Street, Tallahassee, FL 32306, USA.
| | - Rebecca A Buchwalter
- Department of Biomedical Sciences, Florida State University, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Ling-Rong Kao
- Department of Biomedical Sciences, Florida State University, 1115 West Call Street, Tallahassee, FL 32306, USA
| | - Timothy L Megraw
- Department of Biomedical Sciences, Florida State University, 1115 West Call Street, Tallahassee, FL 32306, USA.
| |
Collapse
|
208
|
Abstract
The organization of microtubule networks is crucial for controlling chromosome segregation during cell division, for positioning and transport of different organelles, and for cell polarity and morphogenesis. The geometry of microtubule arrays strongly depends on the localization and activity of the sites where microtubules are nucleated and where their minus ends are anchored. Such sites are often clustered into structures known as microtubule-organizing centers, which include the centrosomes in animals and spindle pole bodies in fungi. In addition, other microtubules, as well as membrane compartments such as the cell nucleus, the Golgi apparatus, and the cell cortex, can nucleate, stabilize, and tether microtubule minus ends. These activities depend on microtubule-nucleating factors, such as γ-tubulin-containing complexes and their activators and receptors, and microtubule minus end-stabilizing proteins with their binding partners. Here, we provide an overview of the current knowledge on how such factors work together to control microtubule organization in different systems.
Collapse
Affiliation(s)
- Jingchao Wu
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands; ,
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands; ,
| |
Collapse
|
209
|
Bestul AJ, Yu Z, Unruh JR, Jaspersen SL. Molecular model of fission yeast centrosome assembly determined by superresolution imaging. J Cell Biol 2017; 216:2409-2424. [PMID: 28619713 PMCID: PMC5551712 DOI: 10.1083/jcb.201701041] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/17/2017] [Accepted: 05/10/2017] [Indexed: 01/06/2023] Open
Abstract
Microtubule-organizing centers (MTOCs), known as centrosomes in animals and spindle pole bodies (SPBs) in fungi, are important for the faithful distribution of chromosomes between daughter cells during mitosis as well as for other cellular functions. The cytoplasmic duplication cycle and regulation of the Schizosaccharomyces pombe SPB is analogous to centrosomes, making it an ideal model to study MTOC assembly. Here, we use superresolution structured illumination microscopy with single-particle averaging to localize 14 S. pombe SPB components and regulators, determining both the relationship of proteins to each other within the SPB and how each protein is assembled into a new structure during SPB duplication. These data enabled us to build the first comprehensive molecular model of the S. pombe SPB, resulting in structural and functional insights not ascertained through investigations of individual subunits, including functional similarities between Ppc89 and the budding yeast SPB scaffold Spc42, distribution of Sad1 to a ring-like structure and multiple modes of Mto1 recruitment.
Collapse
Affiliation(s)
| | - Zulin Yu
- Stowers Institute for Medical Research, Kansas City, MO
| | - Jay R Unruh
- Stowers Institute for Medical Research, Kansas City, MO
| | - Sue L Jaspersen
- Stowers Institute for Medical Research, Kansas City, MO .,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
210
|
Sulimenko V, Hájková Z, Klebanovych A, Dráber P. Regulation of microtubule nucleation mediated by γ-tubulin complexes. PROTOPLASMA 2017; 254:1187-1199. [PMID: 28074286 DOI: 10.1007/s00709-016-1070-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/22/2016] [Indexed: 05/18/2023]
Abstract
The microtubule cytoskeleton is critically important for spatio-temporal organization of eukaryotic cells. The nucleation of new microtubules is typically restricted to microtubule organizing centers (MTOCs) and requires γ-tubulin that assembles into multisubunit complexes of various sizes. γ-Tubulin ring complexes (TuRCs) are efficient microtubule nucleators and are associated with large number of targeting, activating and modulating proteins. γ-Tubulin-dependent nucleation of microtubules occurs both from canonical MTOCs, such as spindle pole bodies and centrosomes, and additional sites such as Golgi apparatus, nuclear envelope, plasma membrane-associated sites, chromatin and surface of pre-existing microtubules. Despite many advances in structure of γ-tubulin complexes and characterization of γTuRC interacting factors, regulatory mechanisms of microtubule nucleation are not fully understood. Here, we review recent work on the factors and regulatory mechanisms that are involved in centrosomal and non-centrosomal microtubule nucleation.
Collapse
Affiliation(s)
- Vadym Sulimenko
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Zuzana Hájková
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Anastasiya Klebanovych
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| | - Pavel Dráber
- Department of Biology of Cytoskeleton, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20, Prague 4, Czech Republic.
| |
Collapse
|
211
|
Chen Y, Bi H, Li X, Zhang Z, Yue H, Weng S, He J. Wsv023 interacted with Litopenaeus vannamei γ-tubulin complex associated proteins 2, and decreased the formation of microtubules. ROYAL SOCIETY OPEN SCIENCE 2017; 4:160379. [PMID: 28484601 PMCID: PMC5414238 DOI: 10.1098/rsos.160379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 03/23/2017] [Indexed: 06/07/2023]
Abstract
A previous study found the key transcription factor of Litopenaeus vannamei PERK-eIF2α pathway cyclic AMP-dependent transcription factor 4 (LvATF4) was involved in the transcriptional regulation of white spot syndrome virus (WSSV) gene wsv023. Knocked-down expression of LvATF4 reduced the viral copy number and the cumulative mortality of WSSV-infected shrimp. These results suggested that wsv023 may be critical to WSSV infection but the precise function of wsv023 was still unknown. By using co-immunoprecipitation and pull-down assays, we show that wsv023 interacts with L. vannamei gamma complex-associated protein 2 (LvGCP2), which is the core protein of the γ-tubulin small complex. Knocked-down, the wsv023 gene significantly reduced the copy number of WSSV in L. vannamei muscle, as well as the cumulative mortality of infected shrimp. And PERK-eIF2α pathway inhibition also showed reduced virus copy number and abrogated shrimp mortality. Furthermore, overexpression of wsv023 inhibited the formation of microtubules in 293T cells. Flow cytometry revealed that WSSV infection similarly decreased the formation of microtubules in L. vannamei haemocytes. These findings suggested that wsv023 plays a role in microtubule organization in host cells, which in turn may be beneficial to WSSV.
Collapse
Affiliation(s)
- Yihong Chen
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, South China Sea Bio-Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), School of Marine Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
- State Key Laboratory for Biocontrol, OE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
| | - Haitao Bi
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
- State Key Laboratory for Biocontrol, OE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
| | - Xiaoyun Li
- Fisheries College, Guangdong Ocean University, Zhanjiang, People's Republic of China
| | - Zezhi Zhang
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
- State Key Laboratory for Biocontrol, OE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
| | - Haitao Yue
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
- State Key Laboratory for Biocontrol, OE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
| | - Shaoping Weng
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
- State Key Laboratory for Biocontrol, OE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
| | - Jianguo He
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, South China Sea Bio-Resource Exploitation and Protection Collaborative Innovation Center (SCS-REPIC), School of Marine Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
- School of Life Sciences, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
- State Key Laboratory for Biocontrol, OE Key Laboratory of Aquatic Product Safety, Institute of Aquatic Economic Animals and Guangdong Province Key Laboratory for Aquatic Economic Animals, Sun Yat-sen University, 135 Xingang Road West, Guangzhou 510275, People's Republic of China
| |
Collapse
|
212
|
Abstract
The mitotic spindle has a crucial role in ensuring the accurate segregation of chromosomes into the two daughter cells during cell division, which is paramount for maintaining genome integrity. It is a self-organized and dynamic macromolecular structure that is constructed from microtubules, microtubule-associated proteins and motor proteins. Thirty years of research have led to the identification of centrosome-, chromatin- and microtubule-mediated microtubule nucleation pathways that each contribute to mitotic spindle assembly. Far from being redundant pathways, data are now emerging regarding how they function together to ensure the timely completion of mitosis. We are also beginning to comprehend the multiple mechanisms by which cells regulate spindle scaling. Together, this research has increased our understanding of how cells coordinate hundreds of proteins to assemble the dynamic, precise and robust structure that is the mitotic spindle.
Collapse
|
213
|
Kapoor TM. Metaphase Spindle Assembly. BIOLOGY 2017; 6:biology6010008. [PMID: 28165376 PMCID: PMC5372001 DOI: 10.3390/biology6010008] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 01/31/2023]
Abstract
A microtubule-based bipolar spindle is required for error-free chromosome segregation during cell division. In this review I discuss the molecular mechanisms required for the assembly of this dynamic micrometer-scale structure in animal cells.
Collapse
Affiliation(s)
- Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology, the Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
214
|
Dráberová E, Sulimenko V, Vinopal S, Sulimenko T, Sládková V, D'Agostino L, Sobol M, Hozák P, Křen L, Katsetos CD, Dráber P. Differential expression of human γ-tubulin isotypes during neuronal development and oxidative stress points to a γ-tubulin-2 prosurvival function. FASEB J 2017; 31:1828-1846. [PMID: 28119396 DOI: 10.1096/fj.201600846rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 01/03/2017] [Indexed: 12/11/2022]
Abstract
γ-Tubulins are highly conserved members of the tubulin superfamily essential for microtubule nucleation. Humans possess 2 γ-tubulin genes. It is thought that γ-tubulin-1 represents a ubiquitous isotype, whereas γ-tubulin-2 is found predominantly in the brain, where it may be endowed with divergent functions beyond microtubule nucleation. The molecular basis of the purported functional differences between γ-tubulins is unknown. We report discrimination of human γ-tubulins according to their electrophoretic and immunochemical properties. In vitro mutagenesis revealed that the differences in electrophoretic mobility originate in the C-terminal regions of the γ-tubulins. Using epitope mapping, we discovered mouse monoclonal antibodies that can discriminate between human γ-tubulin isotypes. Real time quantitative RT-PCR and 2-dimensional-PAGE showed that γ-tubulin-1 is the dominant isotype in fetal neurons. Although γ-tubulin-2 accumulates in the adult brain, γ-tubulin-1 remains the major isotype in various brain regions. Localization of γ-tubulin-1 in mature neurons was confirmed by immunohistochemistry and immunofluorescence microscopy on clinical samples and tissue microarrays. Differentiation of SH-SY5Y human neuroblastoma cells by all-trans retinoic acid, or oxidative stress induced by mitochondrial inhibitors, resulted in upregulation of γ-tubulin-2, whereas the expression of γ-tubulin-1 was unchanged. Fractionation experiments and immunoelectron microscopy revealed an association of γ-tubulins with mitochondrial membranes. These data indicate that in the face of predominant γ-tubulin-1 expression, the accumulation of γ-tubulin-2 in mature neurons and neuroblastoma cells during oxidative stress may denote a prosurvival role of γ-tubulin-2 in neurons.-Dráberová, E., Sulimenko, V., Vinopal, S., Sulimenko, T., Sládková, V., D'Agostino, L., Sobol, M., Hozák, P., Křen, L., Katsetos, C. D., Dráber, P. Differential expression of human γ-tubulin isotypes during neuronal development and oxidative stress points to γ-tubulin-2 prosurvival function.
Collapse
Affiliation(s)
- Eduarda Dráberová
- Department of Biology of Cytoskeleton, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Vadym Sulimenko
- Department of Biology of Cytoskeleton, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Stanislav Vinopal
- Department of Biology of Cytoskeleton, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Tetyana Sulimenko
- Department of Biology of Cytoskeleton, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Vladimíra Sládková
- Department of Biology of Cytoskeleton, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Luca D'Agostino
- Department of Pediatrics, Drexel University College of Medicine, St. Christopher's Hospital for Children and Hahnemann University Hospital, Philadelphia, Pennsylvania, USA
| | - Margaryta Sobol
- Department of the Nucleus, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Pavel Hozák
- Department of the Nucleus, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Leoš Křen
- Department of Pathology and Laboratory Medicine, Drexel University College of Medicine, St. Christopher's Hospital for Children and Hahnemann University Hospital, Philadelphia, Pennsylvania, USA; and
| | - Christos D Katsetos
- Department of Pediatrics, Drexel University College of Medicine, St. Christopher's Hospital for Children and Hahnemann University Hospital, Philadelphia, Pennsylvania, USA
| | - Pavel Dráber
- Department of Biology of Cytoskeleton, Academy of Sciences of the Czech Republic, Prague, Czech Republic;
| |
Collapse
|
215
|
Mohammed AM, Velazquez L, Chisenhall A, Schiffels D, Fygenson DK, Schulman R. Self-assembly of precisely defined DNA nanotube superstructures using DNA origami seeds. NANOSCALE 2017; 9:522-526. [PMID: 27957574 DOI: 10.1039/c6nr06983e] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
We demonstrate a versatile process for assembling micron-scale filament architectures by controlling where DNA tile nanotubes nucleate on DNA origami assemblies. "Nunchucks," potential mechanical magnifiers of nanoscale dynamics consisting of two nanotubes connected by a dsDNA linker, form at yields sufficient for application and consistent with models.
Collapse
Affiliation(s)
- A M Mohammed
- Chemical and Biomolecular Engineering, Johns Hopkins University, USA.
| | - L Velazquez
- Department of Physics, University of California Santa Barbara, USA
| | - A Chisenhall
- Chemical and Biomolecular Engineering, Johns Hopkins University, USA.
| | - D Schiffels
- Department of Physics, University of California Santa Barbara, USA
| | - D K Fygenson
- Department of Physics, University of California Santa Barbara, USA and Biomolecular Science and Engineering, University of California Santa Barbara, USA
| | - R Schulman
- Chemical and Biomolecular Engineering, Johns Hopkins University, USA. and Computer Science, Johns Hopkins University, USA
| |
Collapse
|
216
|
Lim NR, Yeap YYC, Ang CS, Williamson NA, Bogoyevitch MA, Quinn LM, Ng DCH. Aurora A phosphorylation of WD40-repeat protein 62 in mitotic spindle regulation. Cell Cycle 2016; 15:413-24. [PMID: 26713495 DOI: 10.1080/15384101.2015.1127472] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Mitotic spindle organization is regulated by centrosomal kinases that potentiate recruitment of spindle-associated proteins required for normal mitotic progress including the microcephaly protein WD40-repeat protein 62 (WDR62). WDR62 functions underlie normal brain development as autosomal recessive mutations and wdr62 loss cause microcephaly. Here we investigate the signaling interactions between WDR62 and the mitotic kinase Aurora A (AURKA) that has been recently shown to cooperate to control brain size in mice. The spindle recruitment of WDR62 is closely correlated with increased levels of AURKA following mitotic entry. We showed that depletion of TPX2 attenuated WDR62 localization at spindle poles indicating that TPX2 co-activation of AURKA is required to recruit WDR62 to the spindle. We demonstrated that AURKA activity contributed to the mitotic phosphorylation of WDR62 residues Ser49 and Thr50 and phosphorylation of WDR62 N-terminal residues was required for spindle organization and metaphase chromosome alignment. Our analysis of several MCPH-associated WDR62 mutants (V65M, R438H and V1314RfsX18) that are mislocalized in mitosis revealed that their interactions and phosphorylation by AURKA was substantially reduced consistent with the notion that AURKA is a key determinant of WDR62 spindle recruitment. Thus, our study highlights the role of AURKA signaling in the spatiotemporal control of WDR62 at spindle poles where it maintains spindle organization.
Collapse
Affiliation(s)
- Nicholas R Lim
- a Department of Biochemistry and Molecular Biology , University of Melbourne , Victoria , Australia.,b Bio21 Molecular Science and Biotechnology Institute, University of Melbourne , Victoria , Australia
| | - Yvonne Y C Yeap
- a Department of Biochemistry and Molecular Biology , University of Melbourne , Victoria , Australia.,d School of Biomedical Sciences, University of Queensland , St Lucia , Australia
| | - Ching-Seng Ang
- b Bio21 Molecular Science and Biotechnology Institute, University of Melbourne , Victoria , Australia
| | - Nicholas A Williamson
- b Bio21 Molecular Science and Biotechnology Institute, University of Melbourne , Victoria , Australia
| | - Marie A Bogoyevitch
- a Department of Biochemistry and Molecular Biology , University of Melbourne , Victoria , Australia.,b Bio21 Molecular Science and Biotechnology Institute, University of Melbourne , Victoria , Australia
| | - Leonie M Quinn
- c Department of Anatomy and Neuroscience , University of Melbourne , Victoria , Australia
| | - Dominic C H Ng
- a Department of Biochemistry and Molecular Biology , University of Melbourne , Victoria , Australia.,d School of Biomedical Sciences, University of Queensland , St Lucia , Australia
| |
Collapse
|
217
|
Schummel PH, Gao M, Winter R. Modulation of the Polymerization Kinetics of α/β-Tubulin by Osmolytes and Macromolecular Crowding. Chemphyschem 2016; 18:189-197. [DOI: 10.1002/cphc.201601032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Indexed: 12/30/2022]
Affiliation(s)
- Paul Hendrik Schummel
- Faculty of Chemistry and Chemical Biology, Physical Chemistry-Biophysical Chemistry; TU Dortmund University; Otto-Hahn-Str. 4a 44227 Dortmund Germany
| | - Mimi Gao
- Faculty of Chemistry and Chemical Biology, Physical Chemistry-Biophysical Chemistry; TU Dortmund University; Otto-Hahn-Str. 4a 44227 Dortmund Germany
| | - Roland Winter
- Faculty of Chemistry and Chemical Biology, Physical Chemistry-Biophysical Chemistry; TU Dortmund University; Otto-Hahn-Str. 4a 44227 Dortmund Germany
| |
Collapse
|
218
|
Desikan RS, Barkovich AJ. Malformations of cortical development. Ann Neurol 2016; 80:797-810. [PMID: 27862206 PMCID: PMC5177533 DOI: 10.1002/ana.24793] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/26/2016] [Accepted: 09/26/2016] [Indexed: 01/05/2023]
Abstract
Malformations of cortical development (MCDs) compose a diverse range of disorders that are common causes of neurodevelopmental delay and epilepsy. With improved imaging and genetic methodologies, the underlying molecular and pathobiological characteristics of several MCDs have been recently elucidated. In this review, we discuss genetic and molecular alterations that disrupt normal cortical development, with emphasis on recent discoveries, and provide detailed radiological features of the most common and important MCDs. Ann Neurol 2016;80:797-810.
Collapse
Affiliation(s)
- Rahul S. Desikan
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - A. James Barkovich
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
219
|
Fang CT, Kuo HH, Pan TS, Yu FC, Yih LH. HSP70 regulates the function of mitotic centrosomes. Cell Mol Life Sci 2016; 73:3949-60. [PMID: 27137183 PMCID: PMC11108311 DOI: 10.1007/s00018-016-2236-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 04/16/2016] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
Abstract
To establish a functional bipolar mitotic spindle, the centrosome expands and matures, acquiring enhanced activities for microtubule (MT) nucleation and assembly at the onset of mitosis. However, the regulatory mechanisms of centrosome maturation and MT assembly from the matured centrosome are largely unknown. In this study, we showed that heat shock protein (HSP) 70 considerably accumulates at the mitotic centrosome during prometaphase to metaphase and is required for bipolar spindle assembly. Inhibition or depletion of HSP70 impaired the function of mitotic centrosome and disrupted MT nucleation and polymerization from the spindle pole, and may thus result in formation of abnormal mitotic spindles. In addition, HSP70 may associate with NEDD1 and γ-tubulin, two pericentriolar material (PCM) components essential for centrosome maturation and MT nucleation. Loss of HSP70 function disrupted the interaction between NEDD1 and γ-tubulin, and reduced their accumulation at the mitotic centrosome. Our results thus demonstrate a role for HSP70 in regulating centrosome integrity during mitosis, and indicate that HSP70 is required for the maintenance of a functional mitotic centrosome that supports the assembly of a bipolar mitotic spindle.
Collapse
Affiliation(s)
- Chieh-Ting Fang
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Hsiao-Hui Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Tiffany S Pan
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Fu-Chi Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan
| | - Ling-Huei Yih
- Department of Life Science, National Taiwan University, Taipei, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
220
|
Rosselló CA, Lindström L, Glindre J, Eklund G, Alvarado-Kristensson M. Gamma-tubulin coordinates nuclear envelope assembly around chromatin. Heliyon 2016; 2:e00166. [PMID: 27699285 PMCID: PMC5037270 DOI: 10.1016/j.heliyon.2016.e00166] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 07/10/2016] [Accepted: 09/16/2016] [Indexed: 10/29/2022] Open
Abstract
The cytosolic role of γ-tubulin as a microtubule organizer has been studied thoroughly, but its nuclear function is poorly understood. Here, we show that γ-tubulin is located throughout the chromatin of demembranated Xenopus laevis sperm and, as the nucleus is formed, γ-tubulin recruits lamin B3 and nuclear membranes. Immunodepletion of γ-tubulin impairs X. laevis assembly of both the lamina and the nuclear membrane. During nuclear formation in mammalian cell lines, γ-tubulin establishes a cellular protein boundary around chromatin that coordinates nuclear assembly of the daughter nuclei. Furthermore, expression of a γ-tubulin mutant that lacks the DNA-binding domain forms chromatin-empty nuclear like structures and demonstrate that a constant interplay between the chromatin-associated and the cytosolic pools of γ-tubulin is required and, when the balance between pools is impaired, aberrant nuclei are formed. We therefore propose that the nuclear protein meshwork formed by γ-tubulin around chromatin coordinates nuclear formation in eukaryotic cells.
Collapse
Affiliation(s)
- Catalina Ana Rosselló
- Division of Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, 20502. Sweden
| | - Lisa Lindström
- Division of Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, 20502. Sweden
| | - Johan Glindre
- Division of Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, 20502. Sweden
| | - Greta Eklund
- Division of Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, 20502. Sweden
| | - Maria Alvarado-Kristensson
- Division of Molecular Pathology, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, 20502. Sweden
| |
Collapse
|
221
|
Farache D, Jauneau A, Chemin C, Chartrain M, Rémy MH, Merdes A, Haren L. Functional Analysis of γ-Tubulin Complex Proteins Indicates Specific Lateral Association via Their N-terminal Domains. J Biol Chem 2016; 291:23112-23125. [PMID: 27660388 DOI: 10.1074/jbc.m116.744862] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Indexed: 11/06/2022] Open
Abstract
Microtubules are nucleated from multiprotein complexes containing γ-tubulin and associated γ-tubulin complex proteins (GCPs). Small complexes (γTuSCs) comprise two molecules of γ-tubulin bound to the C-terminal domains of GCP2 and GCP3. γTuSCs associate laterally into helical structures, providing a structural template for microtubule nucleation. In most eukaryotes γTuSCs associate with additional GCPs (4, 5, and 6) to form the core of the so-called γ-tubulin ring complex (γTuRC). GCPs 2-6 constitute a family of homologous proteins. Previous structural analysis and modeling of GCPs suggest that all family members can potentially integrate into the helical structure. Here we provide experimental evidence for this model. Using chimeric proteins in which the N- and C-terminal domains of different GCPs are swapped, we show that the N-terminal domains define the functional identity of GCPs, whereas the C-terminal domains are exchangeable. FLIM-FRET experiments indicate that GCP4 and GCP5 associate laterally within the complex, and their interaction is mediated by their N-terminal domains as previously shown for γTuSCs. Our results suggest that all GCPs are incorporated into the helix via lateral interactions between their N-terminal domains, whereas the C-terminal domains mediate longitudinal interactions with γ-tubulin. Moreover, we show that binding to γ-tubulin is not essential for integrating into the helical complex.
Collapse
Affiliation(s)
- Dorian Farache
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Alain Jauneau
- Plateforme Imagerie-Microscopie, FR 3450 Pôle de Biotechnologie Végétale, 31326 Castanet-Tolosan, France
| | - Cécile Chemin
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Marine Chartrain
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Marie-Hélène Rémy
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Andreas Merdes
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| | - Laurence Haren
- From the Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France and
| |
Collapse
|
222
|
Bauer M, Cubizolles F, Schmidt A, Nigg EA. Quantitative analysis of human centrosome architecture by targeted proteomics and fluorescence imaging. EMBO J 2016; 35:2152-2166. [PMID: 27539480 PMCID: PMC5048348 DOI: 10.15252/embj.201694462] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/25/2016] [Indexed: 12/14/2022] Open
Abstract
Centrioles are essential for the formation of centrosomes and cilia. While numerical and/or structural centrosomes aberrations are implicated in cancer, mutations in centriolar and centrosomal proteins are genetically linked to ciliopathies, microcephaly, and dwarfism. The evolutionarily conserved mechanisms underlying centrosome biogenesis are centered on a set of key proteins, including Plk4, Sas-6, and STIL, whose exact levels are critical to ensure accurate reproduction of centrioles during cell cycle progression. However, neither the intracellular levels of centrosomal proteins nor their stoichiometry within centrosomes is presently known. Here, we have used two complementary approaches, targeted proteomics and EGFP-tagging of centrosomal proteins at endogenous loci, to measure protein abundance in cultured human cells and purified centrosomes. Our results provide a first assessment of the absolute and relative amounts of major components of the human centrosome. Specifically, they predict that human centriolar cartwheels comprise up to 16 stacked hubs and 1 molecule of STIL for every dimer of Sas-6. This type of quantitative information will help guide future studies of the molecular basis of centrosome assembly and function.
Collapse
Affiliation(s)
- Manuel Bauer
- Biozentrum, University of Basel, Basel, Switzerland
| | | | | | - Erich A Nigg
- Biozentrum, University of Basel, Basel, Switzerland
| |
Collapse
|
223
|
Kwon HJ, Park JE, Song H, Jang CY. DDA3 and Mdp3 modulate Kif2a recruitment onto the mitotic spindle to control minus-end spindle dynamics. J Cell Sci 2016; 129:2719-25. [PMID: 27284004 DOI: 10.1242/jcs.180109] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 05/25/2016] [Indexed: 11/20/2022] Open
Abstract
Active turnover of spindle microtubules (MTs) for the formation of a bi-orientated spindle, chromosome congression and proper chromosome segregation is regulated by MT depolymerases such as the kinesin-13 family and the plus-end-tracking proteins (+TIPs). However, the control mechanisms underlying the spindle MT dynamics that are responsible for poleward flux at the minus end of MTs are poorly understood. Here, we show that Mdp3 (also known as MAP7D3) forms a complex with DDA3 (also known as PSRC1) and controls spindle dynamics at the minus end of MTs by inhibiting DDA3-mediated Kif2a recruitment to the spindle. Aberrant Kif2a activity at the minus end of spindle MTs in Mdp3-depleted cells decreased spindle stability and resulted in unaligned chromosomes in metaphase, lagging chromosomes in anaphase, and chromosome bridges in telophase and cytokinesis. Although they play opposing roles in minus-end MT dynamics, acting as an MT destabilizer and an MT stabilizer, respectively, DDA3 and Mdp3 did not affect the localization of each other. Thus, the DDA3 complex orchestrates MT dynamics at the MT minus end by fine-tuning the recruitment of Kif2a to regulate minus-end MT dynamics and poleward MT flux at the mitotic spindle.
Collapse
Affiliation(s)
- Hye Jin Kwon
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Ji Eun Park
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Haiyu Song
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Chang-Young Jang
- Research Center for Cell Fate Control, College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| |
Collapse
|
224
|
Sana TG, Berni B, Bleves S. The T6SSs of Pseudomonas aeruginosa Strain PAO1 and Their Effectors: Beyond Bacterial-Cell Targeting. Front Cell Infect Microbiol 2016; 6:61. [PMID: 27376031 PMCID: PMC4899435 DOI: 10.3389/fcimb.2016.00061] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/23/2016] [Indexed: 02/05/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen responsible for many diseases such as chronic lung colonization in cystic fibrosis patients and acute infections in hospitals. The capacity of P. aeruginosa to be pathogenic toward several hosts is notably due to different secretion systems. Amongst them, P. aeruginosa encodes three Type Six Secretion Systems (T6SS), named H1- to H3-T6SS, that act against either prokaryotes and/or eukaryotic cells. They are independent from each other and inject diverse toxins that interact with different components in the host cell. Here we summarize the roles of these T6SSs in the PAO1 strain, as well as the toxins injected and their targets. While H1-T6SS is only involved in antiprokaryotic activity through at least seven different toxins, H2-T6SS and H3-T6SS are also able to target prokaryotic as well as eukaryotic cells. Moreover, recent studies proposed that H2- and H3-T6SS have a role in epithelial cells invasion by injecting at least three different toxins. The diversity of T6SS effectors is astounding and other effectors still remain to be discovered. In this review, we present a table with other putative P. aeruginosa strain PAO1 T6SS-dependent effectors. Altogether, the T6SSs of P. aeruginosa are important systems that help fight other bacteria for their ecological niche, and are important in the pathogenicity process.
Collapse
Affiliation(s)
- Thibault G Sana
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (UMR7255), IMM, Centre National de la Recherche Scientifique and Aix-Marseille UniversityMarseille, France; Department of Microbiology and Immunology, Stanford School of Medicine, Stanford UniversityStanford, CA, USA
| | - Benjamin Berni
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (UMR7255), IMM, Centre National de la Recherche Scientifique and Aix-Marseille University Marseille, France
| | - Sophie Bleves
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (UMR7255), IMM, Centre National de la Recherche Scientifique and Aix-Marseille University Marseille, France
| |
Collapse
|
225
|
14-3-3γ Prevents Centrosome Amplification and Neoplastic Progression. Sci Rep 2016; 6:26580. [PMID: 27253419 PMCID: PMC4890593 DOI: 10.1038/srep26580] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/04/2016] [Indexed: 12/21/2022] Open
Abstract
More than 80% of malignant tumors show centrosome amplification and clustering. Centrosome amplification results from aberrations in the centrosome duplication cycle, which is strictly coordinated with DNA-replication-cycle. However, the relationship between cell-cycle regulators and centrosome duplicating factors is not well understood. This report demonstrates that 14-3-3γ localizes to the centrosome and 14-3-3γ loss leads to centrosome amplification. Loss of 14-3-3γ results in the phosphorylation of NPM1 at Thr-199, causing early centriole disjunction and centrosome hyper-duplication. The centrosome amplification led to aneuploidy and increased tumor formation in mice. Importantly, an increase in passage of the 14-3-3γ-knockdown cells led to an increase in the number of cells containing clustered centrosomes leading to the generation of pseudo-bipolar spindles. The increase in pseudo-bipolar spindles was reversed and an increase in the number of multi-polar spindles was observed upon expression of a constitutively active 14-3-3-binding-defective-mutant of cdc25C (S216A) in the 14-3-3γ knockdown cells. The increase in multi-polar spindle formation was associated with decreased cell viability and a decrease in tumor growth. Our findings uncover the molecular basis of regulation of centrosome duplication by 14-3-3γ and inhibition of tumor growth by premature activation of the mitotic program and the disruption of centrosome clustering.
Collapse
|
226
|
Masuda H, Toda T. Synergistic role of fission yeast Alp16GCP6 and Mzt1MOZART1 in γ-tubulin complex recruitment to mitotic spindle pole bodies and spindle assembly. Mol Biol Cell 2016; 27:1753-63. [PMID: 27053664 PMCID: PMC4884066 DOI: 10.1091/mbc.e15-08-0577] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 03/23/2016] [Accepted: 03/29/2016] [Indexed: 11/30/2022] Open
Abstract
In fission yeast, γ-tubulin ring complex (γTuRC)-specific components Gfh1(GCP4), Mod21(GCP5), and Alp16(GCP6) are nonessential for cell growth. Of these deletion mutants, only alp16Δ shows synthetic lethality with temperature-sensitive mutants of Mzt1(MOZART1), a component of the γTuRC required for recruitment of the complex to microtubule-organizing centers. γ-Tubulin small complex levels at mitotic spindle pole bodies (SPBs, the centrosome equivalent in fungi) and microtubule levels for preanaphase spindles are significantly reduced in alp16Δ cells but not in gfh1Δ or mod21Δ cells. Furthermore, alp16Δ cells often form monopolar spindles and frequently lose a minichromosome when the spindle assembly checkpoint is inactivated. Alp16(GCP6) promotes Mzt1-dependent γTuRC recruitment to mitotic SPBs and enhances spindle microtubule assembly in a manner dependent on its expression levels. Gfh1(GCP4) and Mod21(GCP5) are not required for Alp16(GCP6)-dependent γTuRC recruitment. Mzt1 has an additional role in the activation of the γTuRC for spindle microtubule assembly. The ratio of Mzt1 to γTuRC levels for preanaphase spindles is higher than at other stages of the cell cycle. Mzt1 overproduction enhances spindle microtubule assembly without affecting γTuRC levels at mitotic SPBs. We propose that Alp16(GCP6) and Mzt1 act synergistically for efficient bipolar spindle assembly to ensure faithful chromosome segregation.
Collapse
Affiliation(s)
- Hirohisa Masuda
- Lincoln's Inn Fields Laboratory, The Francis Crick Institute, London WC2A 3LY, United Kingdom
| | - Takashi Toda
- Lincoln's Inn Fields Laboratory, The Francis Crick Institute, London WC2A 3LY, United Kingdom Hiroshima Research Center for Healthy Aging, Department of Molecular Biotechnology, Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima 739-8530, Japan
| |
Collapse
|
227
|
Lyon AS, Morin G, Moritz M, Yabut KCB, Vojnar T, Zelter A, Muller E, Davis TN, Agard DA. Higher-order oligomerization of Spc110p drives γ-tubulin ring complex assembly. Mol Biol Cell 2016; 27:2245-58. [PMID: 27226487 PMCID: PMC4945142 DOI: 10.1091/mbc.e16-02-0072] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/19/2016] [Indexed: 02/01/2023] Open
Abstract
Assembly of the microtubule-nucleating γ-tubulin ring complex (γTuRC) requires higher-order oligomerization of Spc110p, which connects γTuRC to the yeast spindle pole body (SPB). Because Spc110p is highly concentrated at the SPB, spatial regulation of microtubule nucleation is thus achieved by exclusive assembly of γTuRCs proximal to the SPB. The microtubule (MT) cytoskeleton plays important roles in many cellular processes. In vivo, MT nucleation is controlled by the γ-tubulin ring complex (γTuRC), a 2.1-MDa complex composed of γ-tubulin small complex (γTuSC) subunits. The mechanisms underlying the assembly of γTuRC are largely unknown. In yeast, the conserved protein Spc110p both stimulates the assembly of the γTuRC and anchors the γTuRC to the spindle pole body. Using a quantitative in vitro FRET assay, we show that γTuRC assembly is critically dependent on the oligomerization state of Spc110p, with higher-order oligomers dramatically enhancing the stability of assembled γTuRCs. Our in vitro findings were confirmed with a novel in vivo γTuSC recruitment assay. We conclude that precise spatial control over MT nucleation is achieved by coupling localization and higher-order oligomerization of the receptor for γTuRC.
Collapse
Affiliation(s)
- Andrew S Lyon
- Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158
| | - Geneviève Morin
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Michelle Moritz
- Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158
| | | | - Tamira Vojnar
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Alex Zelter
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Eric Muller
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Trisha N Davis
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - David A Agard
- Department of Biochemistry and Biophysics and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
228
|
Piri N, Kwong JMK, Gu L, Caprioli J. Heat shock proteins in the retina: Focus on HSP70 and alpha crystallins in ganglion cell survival. Prog Retin Eye Res 2016; 52:22-46. [PMID: 27017896 PMCID: PMC4842330 DOI: 10.1016/j.preteyeres.2016.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 03/14/2016] [Accepted: 03/22/2016] [Indexed: 10/22/2022]
Abstract
Heat shock proteins (HSPs) belong to a superfamily of stress proteins that are critical constituents of a complex defense mechanism that enhances cell survival under adverse environmental conditions. Cell protective roles of HSPs are related to their chaperone functions, antiapoptotic and antinecrotic effects. HSPs' anti-apoptotic and cytoprotective characteristics, their ability to protect cells from a variety of stressful stimuli, and the possibility of their pharmacological induction in cells under pathological stress make these proteins an attractive therapeutic target for various neurodegenerative diseases; these include Alzheimer's, Parkinson's, Huntington's, prion disease, and others. This review discusses the possible roles of HSPs, particularly HSP70 and small HSPs (alpha A and alpha B crystallins) in enhancing the survival of retinal ganglion cells (RGCs) in optic neuropathies such as glaucoma, which is characterized by progressive loss of vision caused by degeneration of RGCs and their axons in the optic nerve. Studies in animal models of RGC degeneration induced by ocular hypertension, optic nerve crush and axotomy show that upregulation of HSP70 expression by hyperthermia, zinc, geranyl-geranyl acetone, 17-AAG (a HSP90 inhibitor), or through transfection of retinal cells with AAV2-HSP70 effectively supports the survival of injured RGCs. RGCs survival was also stimulated by overexpression of alpha A and alpha B crystallins. These findings provide support for translating the HSP70- and alpha crystallin-based cell survival strategy into therapy to protect and rescue injured RGCs from degeneration associated with glaucomatous and other optic neuropathies.
Collapse
Affiliation(s)
- Natik Piri
- Stein Eye Institute, University of California, Los Angeles, CA 90095, USA; Brain Research Institute, University of California, Los Angeles, CA 90095, USA.
| | - Jacky M K Kwong
- Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Lei Gu
- Stein Eye Institute, University of California, Los Angeles, CA 90095, USA
| | - Joseph Caprioli
- Stein Eye Institute, University of California, Los Angeles, CA 90095, USA; Brain Research Institute, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
229
|
Abstract
Dendrite branching is an essential process for building complex nervous systems. It determines the number, distribution and integration of inputs into a neuron, and is regulated to create the diverse dendrite arbor branching patterns characteristic of different neuron types. The microtubule cytoskeleton is critical to provide structure and exert force during dendrite branching. It also supports the functional requirements of dendrites, reflected by differential microtubule architectural organization between neuron types, illustrated here for sensory neurons. Both anterograde and retrograde microtubule polymerization occur within growing dendrites, and recent studies indicate that branching is enhanced by anterograde microtubule polymerization events in nascent branches. The polarities of microtubule polymerization events are regulated by the position and orientation of microtubule nucleation events in the dendrite arbor. Golgi outposts are a primary microtubule nucleation center in dendrites and share common nucleation machinery with the centrosome. In addition, pre-existing dendrite microtubules may act as nucleation sites. We discuss how balancing the activities of distinct nucleation machineries within the growing dendrite can alter microtubule polymerization polarity and dendrite branching, and how regulating this balance can generate neuron type-specific morphologies.
Collapse
Affiliation(s)
- Caroline Delandre
- a Laboratory for Genetic Control of Neuronal Architecture, RIKEN Brain Science Institute , Wako , Saitama , Japan
| | - Reiko Amikura
- a Laboratory for Genetic Control of Neuronal Architecture, RIKEN Brain Science Institute , Wako , Saitama , Japan
| | - Adrian W Moore
- a Laboratory for Genetic Control of Neuronal Architecture, RIKEN Brain Science Institute , Wako , Saitama , Japan
| |
Collapse
|
230
|
|
231
|
Garrido G, Vernos I. Non-centrosomal TPX2-Dependent Regulation of the Aurora A Kinase: Functional Implications for Healthy and Pathological Cell Division. Front Oncol 2016; 6:88. [PMID: 27148480 PMCID: PMC4831974 DOI: 10.3389/fonc.2016.00088] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/29/2016] [Indexed: 01/09/2023] Open
Abstract
Aurora A has been extensively characterized as a centrosomal kinase with essential functions during cell division including centrosome maturation and separation and spindle assembly. However, Aurora A localization is not restricted to the centrosomes and compelling evidence support the existence of specific mechanisms of activation and functions for non-centrosomal Aurora A in the dividing cell. It has been now well established that spindle assembly involves an acentrosomal RanGTP-dependent pathway that triggers microtubule assembly and organization in the proximity of the chromosomes whether centrosomes are present or not. The mechanism involves the regulation of a number of NLS-containing proteins, generically called SAFS (Spindle Assembly Factors) that exert their functions upon release from karyopherins by RanGTP. One of them, the nuclear protein TPX2 interacts with and activates Aurora A upon release from importins by RanGTP. This basic mechanism triggers the activation of Aurora A in the proximity of the chromosomes potentially translating the RanGTP signaling gradient centered on the chromosome into an Aurora A phosphorylation network. Here, we will review our current knowledge on the RanGTP-dependent TPX2 activation of Aurora A away from centrosomes: from the mechanism of activation and its functional consequences on the kinase stability and regulation to its roles in spindle assembly and cell division. We will then focus on the substrates of the TPX2-activated Aurora A having a role in microtubule nucleation, stabilization, and organization. Finally, we will briefly discuss the implications of the use of Aurora A inhibitors in anti-tumor therapies in the light of its functional interaction with TPX2.
Collapse
Affiliation(s)
- Georgina Garrido
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Isabelle Vernos
- Cell and Developmental Biology Programme, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
232
|
Ohashi T, Yamamoto T, Yamanashi Y, Ohsugi M. Human TUBG2 gene is expressed as two splice variant mRNA and involved in cell growth. FEBS Lett 2016; 590:1053-63. [PMID: 27015882 DOI: 10.1002/1873-3468.12163] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/08/2016] [Accepted: 03/23/2016] [Indexed: 01/14/2023]
Abstract
In mammals, γ-tubulin, a key protein in microtubule nucleation, is encoded by two genes, TUBG1 and TUBG2. Human TUBG1 and TUBG2 mRNA are expressed ubiquitously and predominantly in preimplantation embryos and the brain, respectively, but specific detection of γ-tubulin2 protein expression is difficult due to their high sequence similarity. Here, we describe a protocol for differential detection of two human γ-tubulins by western blotting. In several cancer cell lines and the brain, expression of γ-tubulin2 along with γ-tubulin1 and a novel TUBG2 splice variant are identified. Contribution of ectopically expressed γ-tubulin2 in cancer growth was determined by depletion of γ-tubulin2.
Collapse
Affiliation(s)
- Tsubasa Ohashi
- Department of Life Sciences Graduate School of Arts and Sciences, The University of Tokyo, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology, Japan.,RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Yuji Yamanashi
- Division of Genetics, Department of Cancer Biology, Institute of Medical Science, University of Tokyo, Japan
| | - Miho Ohsugi
- Department of Life Sciences Graduate School of Arts and Sciences, The University of Tokyo, Japan
| |
Collapse
|
233
|
Ti SC, Pamula MC, Howes SC, Duellberg C, Cade NI, Kleiner RE, Forth S, Surrey T, Nogales E, Kapoor TM. Mutations in Human Tubulin Proximal to the Kinesin-Binding Site Alter Dynamic Instability at Microtubule Plus- and Minus-Ends. Dev Cell 2016; 37:72-84. [PMID: 27046833 PMCID: PMC4832424 DOI: 10.1016/j.devcel.2016.03.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/12/2016] [Accepted: 03/04/2016] [Indexed: 01/10/2023]
Abstract
The assembly of microtubule-based cellular structures depends on regulated tubulin polymerization and directional transport. Here, we purify and characterize tubulin heterodimers that have human β-tubulin isotype III (TUBB3), as well as heterodimers with one of two β-tubulin mutations (D417H or R262H). Both point mutations are proximal to the kinesin-binding site and have been linked to an ocular motility disorder in humans. Compared to wild-type, microtubules with these mutations have decreased catastrophe frequencies and increased average lifetimes of plus- and minus-end-stabilizing caps. Importantly, the D417H mutation does not alter microtubule lattice structure or Mal3 binding to growing filaments. Instead, this mutation reduces the affinity of tubulin for TOG domains and colchicine, suggesting that the distribution of tubulin heterodimer conformations is changed. Together, our findings reveal how residues on the surface of microtubules, distal from the GTP-hydrolysis site and inter-subunit contacts, can alter polymerization dynamics at the plus- and minus-ends of microtubules.
Collapse
Affiliation(s)
- Shih-Chieh Ti
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Melissa C Pamula
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Stuart C Howes
- Biophysics Graduate Group, University of California, Berkeley, CA 94720, USA
| | - Christian Duellberg
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Nicholas I Cade
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Ralph E Kleiner
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Scott Forth
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Thomas Surrey
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Eva Nogales
- Life Sciences Division, Lawrence Berkeley National Lab, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Tarun M Kapoor
- Laboratory of Chemistry and Cell Biology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
234
|
Higa GM, Sypult C. Molecular Biology and Clinical Mitigation of Cancer Treatment-Induced Neuropathy. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2016; 10:27-34. [PMID: 27081324 PMCID: PMC4820064 DOI: 10.4137/cmo.s32810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 02/21/2016] [Accepted: 02/22/2016] [Indexed: 01/12/2023]
Abstract
Disruption of microtubule function is the antitumor mechanism of several classes of drugs used to treat cancer today. However, the significant beneficial effect on tumor outcomes is frequently counterbalanced by neurotoxic complications. Despite an abundance of scientific data, our under-standing of the biological mechanisms underlying this toxic reaction remains unclear, further hindering attempts to identify and develop effective preventive strategies. The primary goals of this review are to: (1) provide insight regarding the biology of the microtubule, (2) analyze the molecular and biochemical pathways that may be involved in the development of neurotoxicity, and (3) propose a unifying concept linking drug-induced neuropathy, microtubule dysfunction, and vitamin D.
Collapse
Affiliation(s)
- Gerald M Higa
- Professor of Clinical Pharmacy, Clinical Professor of Medicine, Schools of Pharmacy and Medicine, West Virginia University, Morgantown, WV, USA
| | - Corbin Sypult
- Doctor of Pharmacy Student, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
235
|
Milunović-Jevtić A, Mooney P, Sulerud T, Bisht J, Gatlin JC. Centrosomal clustering contributes to chromosomal instability and cancer. Curr Opin Biotechnol 2016; 40:113-118. [PMID: 27046071 DOI: 10.1016/j.copbio.2016.03.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/07/2016] [Accepted: 03/15/2016] [Indexed: 12/18/2022]
Abstract
Cells assemble mitotic spindles during each round of division to insure accurate segregation of their duplicated genome. In animal cells, stereotypical spindles have two poles, each containing one centrosome, from which microtubules are nucleated. By contrast, many cancer cells often contain more than two centrosomes and form transient multipolar spindle structures with more than two poles. In order to divide and produce viable progeny, the multipolar spindle intermediate must be reshaped into a pseudo-bipolar structure via a process called centrosomal clustering. Pseudo-bipolar spindles appear to function normally during mitosis, but they occasionally give rise to aneuploid and transformed daughter cells. Agents that inhibit centrosomal clustering might therefore work as a potential cancer therapy, specifically targeting mitosis in supernumerary centrosome-containing cells.
Collapse
Affiliation(s)
| | - P Mooney
- University of Wyoming, Department of Molecular Biology, United States
| | - T Sulerud
- University of Wyoming, Department of Molecular Biology, United States
| | - J Bisht
- University of Wyoming, Department of Molecular Biology, United States
| | - J C Gatlin
- University of Wyoming, Department of Molecular Biology, United States.
| |
Collapse
|
236
|
Cearns MD, Escuin S, Alexandre P, Greene NDE, Copp AJ. Microtubules, polarity and vertebrate neural tube morphogenesis. J Anat 2016; 229:63-74. [PMID: 27025884 DOI: 10.1111/joa.12468] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2016] [Indexed: 12/20/2022] Open
Abstract
Microtubules (MTs) are key cellular components, long known to participate in morphogenetic events that shape the developing embryo. However, the links between the cellular functions of MTs, their effects on cell shape and polarity, and their role in large-scale morphogenesis remain poorly understood. Here, these relationships were examined with respect to two strategies for generating the vertebrate neural tube: bending and closure of the mammalian neural plate; and cavitation of the teleost neural rod. The latter process has been compared with 'secondary' neurulation that generates the caudal spinal cord in mammals. MTs align along the apico-basal axis of the mammalian neuroepithelium early in neural tube closure, participating functionally in interkinetic nuclear migration, which indirectly impacts on cell shape. Whether MTs play other functional roles in mammalian neurulation remains unclear. In the zebrafish, MTs are important for defining the neural rod midline prior to its cavitation, both by localizing apical proteins at the tissue midline and by orienting cell division through a mirror-symmetric MT apparatus that helps to further define the medial localization of apical polarity proteins. Par proteins have been implicated in centrosome positioning in neuroepithelia as well as in the control of polarized morphogenetic movements in the neural rod. Understanding of MT functions during early nervous system development has so far been limited, partly by techniques that fail to distinguish 'cause' from 'effect'. Future developments will likely rely on novel ways to selectively impair MT function in order to investigate the roles they play.
Collapse
Affiliation(s)
- Michael D Cearns
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| | - Sarah Escuin
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| | - Paula Alexandre
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| | - Nicholas D E Greene
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| | - Andrew J Copp
- Newlife Birth Defects Research Centre, Institute of Child Health, University College London, London, UK
| |
Collapse
|
237
|
Abstract
In human cells, the basal body (BB) core comprises a ninefold microtubule-triplet cylindrical structure. Distal and subdistal appendages are located at the distal end of BB, where they play indispensable roles in cilium formation and function. Most cells that arrest in the G0 stage of the cell cycle initiate BB docking at the plasma membrane followed by BB-mediated growth of a solitary primary cilium, a structure required for sensing the extracellular environment and cell signaling. In addition to the primary cilium, motile cilia are present in specialized cells, such as sperm and airway epithelium. Mutations that affect BB function result in cilia dysfunction. This can generate syndromic disorders, collectively called ciliopathies, for which there are no effective treatments. In this review, we focus on the features and functions of BBs and centrosomes in Homo sapiens.
Collapse
Affiliation(s)
- Anastassiia Vertii
- />Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA USA
| | - Hui-Fang Hung
- />Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA USA
| | - Heidi Hehnly
- />Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY USA
| | - Stephen Doxsey
- />Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA USA
| |
Collapse
|
238
|
Abstract
Microtubule dynamics are fundamental for many aspects of cell physiology, but their mechanistic underpinnings remain unclear despite 40 years of intense research. In recent years, the continued union of reconstitution biochemistry, structural biology, and modeling has yielded important discoveries that deepen our understanding of microtubule dynamics. These studies, which we review here, underscore the importance of GTP hydrolysis-induced changes in tubulin structure as microtubules assemble, and highlight the fact that each aspect of microtubule behavior is the output of complex, multi-step processes. Although this body of work moves us closer to appreciating the key features of microtubule biochemistry that drive dynamic instability, the divide between our understanding of microtubules in isolation versus within the cellular milieu remains vast. Bridging this gap will serve as fertile grounds of cytoskeleton-focused research for many years to come.
Collapse
Affiliation(s)
- Ryoma Ohi
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Marija Zanic
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
239
|
Greenberg CH, Kollman J, Zelter A, Johnson R, MacCoss MJ, Davis TN, Agard DA, Sali A. Structure of γ-tubulin small complex based on a cryo-EM map, chemical cross-links, and a remotely related structure. J Struct Biol 2016; 194:303-10. [PMID: 26968363 DOI: 10.1016/j.jsb.2016.03.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/06/2016] [Accepted: 03/07/2016] [Indexed: 11/26/2022]
Abstract
Modeling protein complex structures based on distantly related homologues can be challenging due to poor sequence and structure conservation. Therefore, utilizing even low-resolution experimental data can significantly increase model precision and accuracy. Here, we present models of the two key functional states of the yeast γ-tubulin small complex (γTuSC): one for the low-activity "open" state and another for the higher-activity "closed" state. Both models were computed based on remotely related template structures and cryo-EM density maps at 6.9Å and 8.0Å resolution, respectively. For each state, extensive sampling of alignments and conformations was guided by the fit to the corresponding cryo-EM density map. The resulting good-scoring models formed a tightly clustered ensemble of conformations in most regions. We found significant structural differences between the two states, primarily in the γ-tubulin subunit regions where the microtubule binds. We also report a set of chemical cross-links that were found to be consistent with equilibrium between the open and closed states. The protocols developed here have been incorporated into our open-source Integrative Modeling Platform (IMP) software package (http://integrativemodeling.org), and can therefore be applied to many other systems.
Collapse
Affiliation(s)
- Charles H Greenberg
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California at San Francisco, San Francisco, CA, USA.
| | - Justin Kollman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Alex Zelter
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Richard Johnson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Michael J MacCoss
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Trisha N Davis
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| | - David A Agard
- Department of Biochemistry and Biophysics, University of California at San Francisco, San Francisco, CA, USA; Howard Hughes Medical Institute, University of California at San Francisco, San Francisco, CA, USA.
| | - Andrej Sali
- Department of Bioengineering and Therapeutic Sciences, Department of Pharmaceutical Chemistry, California Institute for Quantitative Biosciences, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
240
|
Mutch CA, Poduri A, Sahin M, Barry B, Walsh CA, Barkovich AJ. Disorders of Microtubule Function in Neurons: Imaging Correlates. AJNR Am J Neuroradiol 2016; 37:528-35. [PMID: 26564436 DOI: 10.3174/ajnr.a4552] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 07/07/2015] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND PURPOSE A number of recent studies have described malformations of cortical development with mutations of components of microtubules and microtubule-associated proteins. Despite examinations of a large number of MRIs, good phenotype-genotype correlations have been elusive. Additionally, most of these studies focused exclusively on cerebral cortical findings. The purpose of this study was to characterize imaging findings associated with disorders of microtubule function. MATERIALS AND METHODS MRIs from 18 patients with confirmed tubulin mutations (8 TUBA1A, 5 TUBB2B, and 5 TUBB3) and 15 patients with known mutations of the genes encoding microtubule-associated proteins (5 LIS1, 4 DCX, and 6 DYNC1H1) were carefully visually analyzed and compared. Specific note was made of the cortical gyral pattern, basal ganglia, and white matter to assess internal capsular size, cortical thickness, ventricular and cisternal size, and the size and contours of the brain stem, cerebellar hemispheres and vermis, and the corpus callosum of patients with tubulin and microtubule-associated protein gene mutations. Results were determined by unanimous consensus of the authors. RESULTS All patients had abnormal findings on MR imaging. A large number of patients with tubulin gene mutations were found to have multiple cortical and subcortical abnormalities, including microcephaly, ventriculomegaly, abnormal gyral and sulcal patterns (termed "dysgyria"), a small or absent corpus callosum, and a small pons. All patients with microtubule-associated protein mutations also had abnormal cerebral cortices (predominantly pachygyria and agyria), but fewer subcortical abnormalities were noted. CONCLUSIONS Comparison of MRIs from patients with known mutations of tubulin genes and microtubule-associated proteins allows the establishment of some early correlations of phenotype with genotype and may assist in identification and diagnosis of these rare disorders.
Collapse
Affiliation(s)
- C A Mutch
- From the Department of Radiology and Biomedical Imaging (C.A.M., A.J.B.), University of California, San Francisco, San Francisco, California
| | - A Poduri
- Epilepsy Genetics Program (A.P., B.B., C.A.W.), Division of Epilepsy and Clinical Neurophysiology F.M. Kirby Neurobiology Center (A.P., B.B., C.A.W.) Division of Genetics and Genomics (B.B., C.A.W.), Department of Medicine, Manton Center for Orphan Disease Research and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts Department of Neurology (A.P., M.S., B.B., C.A.W.), Harvard Medical School, Boston, Massachusetts
| | - M Sahin
- Department of Neurology (A.P., M.S., B.B., C.A.W.), Harvard Medical School, Boston, Massachusetts
| | - B Barry
- Epilepsy Genetics Program (A.P., B.B., C.A.W.), Division of Epilepsy and Clinical Neurophysiology F.M. Kirby Neurobiology Center (A.P., B.B., C.A.W.) Department of Neurology (A.P., M.S., B.B., C.A.W.), Harvard Medical School, Boston, Massachusetts
| | - C A Walsh
- Epilepsy Genetics Program (A.P., B.B., C.A.W.), Division of Epilepsy and Clinical Neurophysiology F.M. Kirby Neurobiology Center (A.P., B.B., C.A.W.) Division of Genetics and Genomics (B.B., C.A.W.), Department of Medicine, Manton Center for Orphan Disease Research and Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts Department of Neurology (A.P., M.S., B.B., C.A.W.), Harvard Medical School, Boston, Massachusetts
| | - A J Barkovich
- From the Department of Radiology and Biomedical Imaging (C.A.M., A.J.B.), University of California, San Francisco, San Francisco, California
| |
Collapse
|
241
|
Dias G, Lino-Neto J, Mercati D, Dallai R. The sperm structure and spermiogenesis of the heteropteran Coptosoma scutellatum (Geoffroy) with emphasis on the development of the centriole adjunct. Micron 2016; 82:33-40. [DOI: 10.1016/j.micron.2015.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/19/2015] [Indexed: 01/07/2023]
|
242
|
Abstract
Microtubules mediate important cellular processes by forming highly ordered arrays. Organization of these networks is achieved by nucleating and anchoring microtubules at centrosomes and other structures collectively known as microtubule-organizing centers (MTOCs). However, the diverse microtubule configurations found in different cell types may not be generated and maintained by MTOCs alone. Work over the last few years has revealed a mechanism that has the capacity to generate cell-type-specific microtubule arrays independently of a specific organizer: nucleation of microtubules from the lateral surface of pre-existing microtubules. This type of nucleation requires cooperation between two different multi-subunit protein complexes, augmin and the γ-tubulin ring complex (γTuRC). Here we review recent molecular insight into microtubule-dependent nucleation and discuss the possibility that the augmin-γTuRC module, initially described in mitosis, may broadly contribute to microtubule organization also in non-mitotic cells.
Collapse
Affiliation(s)
- Carlos Sánchez-Huertas
- Institute for Research in Biomedicine (IRB Barcelona), C/ Baldiri Reixac 10, Barcelona, Spain
| | - Jens Lüders
- Institute for Research in Biomedicine (IRB Barcelona), C/ Baldiri Reixac 10, Barcelona, Spain.
| |
Collapse
|
243
|
Lecland N, Lüders J. Imaging and Quantifying the Dynamics of γ-Tubulin at Microtubule Minus Ends in Mitotic Spindles. Methods Mol Biol 2016; 1413:63-75. [PMID: 27193843 DOI: 10.1007/978-1-4939-3542-0_5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Understanding the organization of complex microtubule arrays such as the mitotic spindle requires information about the position and dynamics of microtubule plus and minus ends. Whereas plus end dynamics have been widely studied using markers such as EB1-GFP, much less is known about the dynamic properties of minus ends, in part because a suitable marker has only recently become available. Here we describe the use of photoactivatable γ-tubulin-paGFP to image and quantify the dynamics of microtubule minus ends in mitotic spindles.
Collapse
Affiliation(s)
- Nicolas Lecland
- Centre Biologie du Développement, UMR 5547 CNRS-Université Paul Sabatier, Toulouse, France
| | - Jens Lüders
- Cell and Developmental Biology Programme, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Baldiri Reixac 10, Barcelona, 08028, Spain.
| |
Collapse
|
244
|
Cota RR, Teixidó-Travesa N, Ezquerra A, Eibes S, Lacasa C, Roig J, Lüders J. MZT1 regulates microtubule nucleation by linking γTuRC assembly to adapter-mediated targeting and activation. J Cell Sci 2016; 130:406-419. [DOI: 10.1242/jcs.195321] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 11/09/2016] [Indexed: 01/22/2023] Open
Abstract
Regulation of the γ-tubulin ring complex (γTuRC) through targeting and activation restricts nucleation of microtubules to microtubule organizing centers (MTOCs), aiding in the assembly of ordered microtubule arrays. However, the mechanistic basis of this important regulation remains poorly understood. Here we show that in human cells γTuRC integrity, determined by the presence of γ-tubulin complex proteins (GCPs) 2-6, is a prerequisite for interaction with the targeting factor NEDD1, impacting on essentially all γ-tubulin dependent functions. Recognition of γTuRC integrity is mediated by MZT1, which binds not only to the GCP3 subunit as previously shown, but cooperatively also to other GCPs through a conserved hydrophobic motif present in the N-termini of GCP2, GCP3, GCP5, and GCP6. MZT1 knockdown causes severe cellular defects under conditions that leave γTuRC intact, suggesting that the essential function of MZT1 is not in γTuRC assembly. Instead, MZT1 specifically binds fully assembled γTuRC to enable interaction with NEDD1 for targeting, and with the CM1 domain of CDK5RAP2 for stimulating nucleation activity. Thus, MZT1 is a ‘priming factor’ for the γTuRC that allows spatial regulation of nucleation.
Collapse
Affiliation(s)
- Rosa Ramírez Cota
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
| | | | - Artur Ezquerra
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
| | - Susana Eibes
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
| | - Cristina Lacasa
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
| | - Joan Roig
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
- Molecular Biology Institute of Barcelona (IBMB-CSIC), 08028 Barcelona, Spain
| | - Jens Lüders
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
| |
Collapse
|
245
|
Guo M, Kim P, Li G, Elowsky C, Alfano J. A Bacterial Effector Co-opts Calmodulin to Target the Plant Microtubule Network. Cell Host Microbe 2016; 19:67-78. [DOI: 10.1016/j.chom.2015.12.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 11/03/2015] [Accepted: 12/21/2015] [Indexed: 12/21/2022]
|
246
|
Toya M, Takeichi M. Organization of Non-centrosomal Microtubules in Epithelial Cells. Cell Struct Funct 2016; 41:127-135. [DOI: 10.1247/csf.16015] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Affiliation(s)
- Mika Toya
- RIKEN Center for Developmental Biology
| | | |
Collapse
|
247
|
Abstract
Microtubules are cytoskeletal filaments that are intrinsically polarized, with two structurally and functionally distinct ends, the plus end and the minus end. Over the last decade, numerous studies have shown that microtubule plus-end dynamics play an important role in many vital cellular processes and are controlled by numerous factors, such as microtubule plus-end-tracking proteins (+TIPs). In contrast, the cellular machinery that controls the behavior and organization of microtubule minus ends remains one of the least well-understood facets of the microtubule cytoskeleton. The recent characterization of the CAMSAP/Patronin/Nezha family members as specific 'minus-end-targeting proteins' ('-TIPs') has provided important new insights into the mechanisms governing minus-end dynamics. Here, we review the current state of knowledge on how microtubule minus ends are controlled and how minus-end regulators contribute to non-centrosomal microtubule organization and function during cell division, migration and differentiation.
Collapse
|
248
|
Colbert PL, Vermeer DW, Wieking BG, Lee JH, Vermeer PD. EphrinB1: novel microtubule associated protein whose expression affects taxane sensitivity. Oncotarget 2015; 6:953-68. [PMID: 25436983 PMCID: PMC4359267 DOI: 10.18632/oncotarget.2823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/25/2014] [Indexed: 11/28/2022] Open
Abstract
Microtubules (MTs) are components of the cytoskeleton made up of polymerized alpha and beta tubulin dimers. MT structure and function must be maintained throughout the cell cycle to ensure proper execution of mitosis and cellular homeostasis. The protein tyrosine phosphatase, PTPN13, localizes to distinct compartments during mitosis and cytokinesis. We have previously demonstrated that the HPV16 E6 oncoprotein binds PTPN13 and leads to its degradation. Thus, we speculated that HPV infection may affect cellular proliferation by altering the localization of a PTPN13 phosphatase substrate, EphrinB1, during mitosis. Here we report that EphrinB1 co-localizes with MTs during all phases of the cell cycle. Specifically, a cleaved, unphosphorylated EphrinB1 fragment directly binds tubulin, while its phosphorylated form lacks MT binding capacity. These findings suggest that EphrinB1 is a novel microtubule associated protein (MAP). Importantly, we show that in the context of HPV16 E6 expression, EphrinB1 affects taxane response in vitro. We speculate that this reflects PTPN13's modulation of EphrinB1 phosphorylation and suggest that EphrinB1 is an important contributor to taxane sensitivity/resistance phenotypes in epithelial cancers. Thus, HPV infection or functional mutations of PTPN13 in non-viral cancers may predict taxane sensitivity.
Collapse
Affiliation(s)
- Paul L Colbert
- Cancer Biology Research Center, Sanford Research, Sioux Falls, South Dakota, USA
| | - Daniel W Vermeer
- Cancer Biology Research Center, Sanford Research, Sioux Falls, South Dakota, USA
| | - Bryant G Wieking
- Cancer Biology Research Center, Sanford Research, Sioux Falls, South Dakota, USA
| | - John H Lee
- Cancer Biology Research Center, Sanford Research, Sioux Falls, South Dakota, USA
| | - Paola D Vermeer
- Cancer Biology Research Center, Sanford Research, Sioux Falls, South Dakota, USA
| |
Collapse
|
249
|
Crespo R, Villar-Alvarez E, Taboada P, Rocha FA, Damas AM, Martins PM. What Can the Kinetics of Amyloid Fibril Formation Tell about Off-pathway Aggregation? J Biol Chem 2015; 291:2018-2032. [PMID: 26601940 DOI: 10.1074/jbc.m115.699348] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Indexed: 11/06/2022] Open
Abstract
Some of the most prevalent neurodegenerative diseases are characterized by the accumulation of amyloid fibrils in organs and tissues. Although the pathogenic role of these fibrils has not been completely established, increasing evidence suggests off-pathway aggregation as a source of toxic/detoxicating deposits that still remains to be targeted. The present work is a step toward the development of off-pathway modulators using the same amyloid-specific dyes as those conventionally employed to screen amyloid inhibitors. We identified a series of kinetic signatures revealing the quantitative importance of off-pathway aggregation relative to amyloid fibrillization; these include non-linear semilog plots of amyloid progress curves, highly variable end point signals, and half-life coordinates weakly influenced by concentration. Molecules that attenuate/intensify the magnitude of these signals are considered promising off-pathway inhibitors/promoters. An illustrative example shows that amyloid deposits of lysozyme are only the tip of an iceberg hiding a crowd of insoluble aggregates. Thoroughly validated using advanced microscopy techniques and complementary measurements of dynamic light scattering, CD, and soluble protein depletion, the new analytical tools are compatible with the high-throughput methods currently employed in drug discovery.
Collapse
Affiliation(s)
- Rosa Crespo
- From the Laboratório de Engenharia de Processos, Ambiente, Biotecnologia e Energia, Departamento de Engenharia Química, Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Eva Villar-Alvarez
- the Departamento de Física de la Materia Condensada, Facultad de Física, Universidad de Santiago de Compostela, 15782 Spain, and
| | - Pablo Taboada
- the Departamento de Física de la Materia Condensada, Facultad de Física, Universidad de Santiago de Compostela, 15782 Spain, and
| | - Fernando A Rocha
- From the Laboratório de Engenharia de Processos, Ambiente, Biotecnologia e Energia, Departamento de Engenharia Química, Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Ana M Damas
- the Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Pedro M Martins
- From the Laboratório de Engenharia de Processos, Ambiente, Biotecnologia e Energia, Departamento de Engenharia Química, Faculdade de Engenharia da Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal,; the Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
250
|
Zrieq R, Sana TG, Vergin S, Garvis S, Volfson I, Bleves S, Voulhoux R, Hegemann JH. Genome-wide Screen of Pseudomonas aeruginosa in Saccharomyces cerevisiae Identifies New Virulence Factors. Front Cell Infect Microbiol 2015; 5:81. [PMID: 26636043 PMCID: PMC4644809 DOI: 10.3389/fcimb.2015.00081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/31/2015] [Indexed: 01/12/2023] Open
Abstract
Pseudomonas aeruginosa is a human opportunistic pathogen that causes mortality in cystic fibrosis and immunocompromised patients. While many virulence factors of this pathogen have already been identified, several remain to be discovered. In this respect we set an unprecedented genome-wide screen of a P. aeruginosa expression library based on a yeast growth phenotype. Fifty-one candidates were selected in athree-round screening process. The robustness of the screen was validated by the selection of three well known secreted proteins including one demonstrated virulence factor, the protease LepA. Further in silico sorting of the 51 candidates highlighted three potential new Pseudomonas effector candidates (Pec). By testing the cytotoxicity of wild type P. aeruginosa vs. pec mutants toward macrophages and the virulence in the Caenorhabditis elegans model, we demonstrated that the three selected Pecs are novel virulence factors of P. aeruginosa. Additional cellular localization experiments in the host revealed specific localization for Pec1 and Pec2 that could inform about their respective functions.
Collapse
Affiliation(s)
- Rafat Zrieq
- Institut für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf Düsseldorf, Germany
| | - Thibault G Sana
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (UMR7255), Institut de Microbiologie de la Méditerranée, Centre National de la Recherche Scientifique, Aix-Marseille Université Marseille, France
| | - Sandra Vergin
- Institut für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf Düsseldorf, Germany
| | - Steve Garvis
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (UMR7255), Institut de Microbiologie de la Méditerranée, Centre National de la Recherche Scientifique, Aix-Marseille Université Marseille, France
| | - Irina Volfson
- Institut für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf Düsseldorf, Germany
| | - Sophie Bleves
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (UMR7255), Institut de Microbiologie de la Méditerranée, Centre National de la Recherche Scientifique, Aix-Marseille Université Marseille, France
| | - Romé Voulhoux
- Laboratoire d'Ingénierie des Systèmes Macromoléculaires (UMR7255), Institut de Microbiologie de la Méditerranée, Centre National de la Recherche Scientifique, Aix-Marseille Université Marseille, France
| | - Johannes H Hegemann
- Institut für Funktionelle Genomforschung der Mikroorganismen, Heinrich-Heine-Universität Düsseldorf Düsseldorf, Germany
| |
Collapse
|