51
|
Ding L, Zhang S, Chen S, Zheng L, Xiao L. Effect and mechanism of lentivirus-mediated silencing of TPX2 gene on proliferation and apoptosis of human hepatoma cells. J Cell Biochem 2019; 120:8352-8358. [PMID: 30548299 DOI: 10.1002/jcb.28119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 10/31/2018] [Indexed: 01/24/2023]
Abstract
This study aimed to investigate the role and mechanism of action of targeting protein for Xklp2 (TPX2) in liver cancer, we compared TPX messenger RNA (mRNA) expression in liver cancer tissue samples and adjacent normal liver tissue samples as well as in human liver cancer cell lines and nonmalignant cell line by quantitative reverse transcription polymerase chain reaction (qRT-PCR). TPX2 gene was silenced in HepG2 cells by transfection with the lentiviral vector expressing TPX2-targeting short hairpin RNA (shRNA), and the knockdown efficiency was evaluated by RT-qPCR. Cell proliferation, apoptosis as well as protein level of c-Myc, cyclin D1, caspase-3, phosphorylated glycogen synthase kinase-3β (p-GSK-3β), and β-catenin in HepG2 cells were evaluated before and after the TPX2 knockdown. Wnt/β-catenin signaling pathway was inhibited by treatment with 20 μM of XAV-939 or activated by treatment with 20 mM of LiCl. We found that TPX2 mRNA level was significantly increased in liver cancer tissue samples and cell lines comparing to noncancerous counterparts (P < 0.05). TPX2 knockdown significantly reduces TPX2 expression (P < 0.01), cell proliferation (P < 0.05), protein level of c-Myc and cyclin D1 (P < 0.01), activation of Wnt/β-catenin signaling in HepG2 cells (P < 0.01) while increasing cell apoptosis (P < 0.01). Treatment with XAV-939 significantly reduced HepG2 cell proliferation (P < 0.05) while increasing cell apoptosis (P < 0.01). Treatment with LiCl significantly attenuated the antiproliferative and apoptosis-promoting effect of TPX2 knockdown on HepG2 cells (P < 0.05). Lentivirus-mediated silencing of TPX2 gene could inhibit proliferation and induce apoptosis in hepatoma cells by inhibiting Wnt signaling pathway and regulating cyclin and apoptosis-related proteins.
Collapse
Affiliation(s)
- Lei Ding
- Department of Infectious Diseases, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Shuhong Zhang
- Department of Infectious Diseases, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Shijun Chen
- Department of infectious diseases, Jinan Infectious Diseases Hospital Affiliated to Shandong University, Jinan, China
| | - Lixue Zheng
- Department of Infectious Diseases, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Lianxiang Xiao
- Department of MRI Diagnosis, Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| |
Collapse
|
52
|
Abstract
Microtubules are major constituents of the cytoskeleton in all eukaryotic cells. They are essential for chromosome segregation during cell division, for directional intracellular transport and for building specialized cellular structures such as cilia or flagella. Their assembly has to be controlled spatially and temporally. For this, the cell uses multiprotein complexes containing γ-tubulin. γ-Tubulin has been found in two different types of complexes, γ-tubulin small complexes and γ-tubulin ring complexes. Binding to adaptors and activator proteins transforms these complexes into structural templates that drive the nucleation of new microtubules in a highly controlled manner. This review discusses recent advances on the mechanisms of assembly, recruitment and activation of γ-tubulin complexes at microtubule-organizing centres.
Collapse
Affiliation(s)
- Dorian Farache
- Centre de Biologie Intégrative, Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France
| | - Laurent Emorine
- Centre de Biologie Intégrative, Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France
| | - Laurence Haren
- Centre de Biologie Intégrative, Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France
| | - Andreas Merdes
- Centre de Biologie Intégrative, Centre de Biologie du Développement, CNRS-Université Toulouse III, 31062 Toulouse, France
| |
Collapse
|
53
|
Abstract
Mitosis is controlled by reversible protein phosphorylation involving specific kinases and phosphatases. A handful of major mitotic protein kinases, such as the cyclin B-CDK1 complex, the Aurora kinases, and Polo-like kinase 1 (PLK1), cooperatively regulate distinct mitotic processes. Research has identified proteins and mechanisms that integrate these kinases into signaling cascades that guide essential mitotic events. These findings have important implications for our understanding of the mechanisms of mitotic regulation and may advance the development of novel antimitotic drugs. We review collected evidence that in vertebrates, the Aurora kinases serve as catalytic subunits of distinct complexes formed with the four scaffold proteins Bora, CEP192, INCENP, and TPX2, which we deem "core" Aurora cofactors. These complexes and the Aurora-PLK1 cascades organized by Bora, CEP192, and INCENP control crucial aspects of mitosis and all pathways of spindle assembly. We compare the mechanisms of Aurora activation in relation to the different spindle assembly pathways and draw a functional analogy between the CEP192 complex and the chromosomal passenger complex that may reflect the coevolution of centrosomes, kinetochores, and the actomyosin cleavage apparatus. We also analyze the roles and mechanisms of Aurora-PLK1 signaling in the cell and centrosome cycles and in the DNA damage response.
Collapse
Affiliation(s)
- Vladimir Joukov
- N.N. Petrov National Medical Research Center of Oncology, Saint-Petersburg 197758, Russian Federation.
| | | |
Collapse
|
54
|
Animal Female Meiosis: The Challenges of Eliminating Centrosomes. Cells 2018; 7:cells7070073. [PMID: 29996518 PMCID: PMC6071224 DOI: 10.3390/cells7070073] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/03/2018] [Accepted: 07/03/2018] [Indexed: 01/02/2023] Open
Abstract
Sexual reproduction requires the generation of gametes, which are highly specialised for fertilisation. Female reproductive cells, oocytes, grow up to large sizes when they accumulate energy stocks and store proteins as well as mRNAs to enable rapid cell divisions after fertilisation. At the same time, metazoan oocytes eliminate their centrosomes, i.e., major microtubule-organizing centres (MTOCs), during or right after the long growth phases. Centrosome elimination poses two key questions: first, how can the centrosome be re-established after fertilisation? In general, metazoan oocytes exploit sperm components, i.e., the basal body of the sperm flagellum, as a platform to reinitiate centrosome production. Second, how do most metazoan oocytes manage to build up meiotic spindles without centrosomes? Oocytes have evolved mechanisms to assemble bipolar spindles solely around their chromosomes without the guidance of pre-formed MTOCs. Female animal meiosis involves microtubule nucleation and organisation into bipolar microtubule arrays in regulated self-assembly under the control of the Ran system and nuclear transport receptors. This review summarises our current understanding of the molecular mechanism underlying self-assembly of meiotic spindles, its spatio-temporal regulation, and the key players governing this process in animal oocytes.
Collapse
|
55
|
Song JG, King MR, Zhang R, Kadzik RS, Thawani A, Petry S. Mechanism of how augmin directly targets the γ-tubulin ring complex to microtubules. J Cell Biol 2018; 217:2417-2428. [PMID: 29875259 PMCID: PMC6028527 DOI: 10.1083/jcb.201711090] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/10/2018] [Accepted: 05/01/2018] [Indexed: 12/28/2022] Open
Abstract
Microtubules (MTs) must be generated from precise locations to form the structural frameworks required for cell shape and function. MTs are nucleated by the γ-tubulin ring complex (γ-TuRC), but it remains unclear how γ-TuRC gets to the right location. Augmin has been suggested to be a γ-TuRC targeting factor and is required for MT nucleation from preexisting MTs. To determine augmin's architecture and function, we purified Xenopus laevis augmin from insect cells. We demonstrate that augmin is sufficient to target γ-TuRC to MTs by in vitro reconstitution. Augmin is composed of two functional parts. One module (tetramer-II) is necessary for MT binding, whereas the other (tetramer-III) interacts with γ-TuRC. Negative-stain electron microscopy reveals that both tetramers fit into the Y-shape of augmin, and MT branching assays reveal that both are necessary for MT nucleation. The finding that augmin can directly bridge MTs with γ-TuRC via these two tetramers adds to our mechanistic understanding of how MTs can be nucleated from preexisting MTs.
Collapse
Affiliation(s)
- Jae-Geun Song
- Department of Molecular Biology, Princeton University, Princeton, NJ
| | - Matthew R King
- Department of Molecular Biology, Princeton University, Princeton, NJ
| | - Rui Zhang
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO
| | - Rachel S Kadzik
- Department of Molecular Biology, Princeton University, Princeton, NJ
| | - Akanksha Thawani
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ
| | - Sabine Petry
- Department of Molecular Biology, Princeton University, Princeton, NJ
| |
Collapse
|
56
|
Abstract
Microtubules are dynamic polymers of αβ-tubulin that are essential for intracellular organization, organelle trafficking and chromosome segregation. Microtubule growth and shrinkage occur via addition and loss of αβ-tubulin subunits, which are biochemical processes. Dynamic microtubules can also engage in mechanical processes, such as exerting forces by pushing or pulling against a load. Recent advances at the intersection of biochemistry and mechanics have revealed the existence of multiple conformations of αβ-tubulin subunits and their central role in dictating the mechanisms of microtubule dynamics and force generation. It has become apparent that microtubule-associated proteins (MAPs) selectively target specific tubulin conformations to regulate microtubule dynamics, and mechanical forces can also influence microtubule dynamics by altering the balance of tubulin conformations. Importantly, the conformational states of tubulin dimers are likely to be coupled throughout the lattice: the conformation of one dimer can influence the conformation of its nearest neighbours, and this effect can propagate over longer distances. This coupling provides a long-range mechanism by which MAPs and forces can modulate microtubule growth and shrinkage. These findings provide evidence that the interplay between biochemistry and mechanics is essential for the cellular functions of microtubules.
Collapse
Affiliation(s)
- Gary J Brouhard
- Department of Biology, McGill University, Montréal, Quebec, Canada.
| | - Luke M Rice
- Department of Biophysics, University of Texas Southwestern, Dallas, TX, USA.
| |
Collapse
|
57
|
Thawani A, Kadzik RS, Petry S. XMAP215 is a microtubule nucleation factor that functions synergistically with the γ-tubulin ring complex. Nat Cell Biol 2018; 20:575-585. [PMID: 29695792 PMCID: PMC5926803 DOI: 10.1038/s41556-018-0091-6] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/20/2018] [Indexed: 12/16/2022]
Abstract
How microtubules (MTs) are generated in the cell is a major question in understanding how the cytoskeleton is assembled. For several decades, γ-tubulin has been accepted as the universal MT nucleator of the cell. Although there is evidence that γ-tubulin complexes are not the sole MT nucleators, identification of other nucleation factors has proven difficult. Here, we report that the well-characterized MT polymerase XMAP215 (chTOG/Msps/Stu2p/Alp14/Dis1 homologue) is essential for MT nucleation in Xenopus egg extracts. The concentration of XMAP215 determines the extent of MT nucleation. Even though XMAP215 and the γ-tubulin ring complex (γ-TuRC) possess minimal nucleation activity individually, together, these factors synergistically stimulate MT nucleation in vitro. The amino-terminal TOG domains 1-5 of XMAP215 bind to αβ-tubulin and promote MT polymerization, whereas the conserved carboxy terminus is required for efficient MT nucleation and directly binds to γ-tubulin. In summary, XMAP215 and γ-TuRC together function as the principal nucleation module that generates MTs in cells.
Collapse
Affiliation(s)
- Akanksha Thawani
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
| | - Rachel S Kadzik
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sabine Petry
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
58
|
Kitaoka M, Heald R, Gibeaux R. Spindle assembly in egg extracts of the Marsabit clawed frog, Xenopus borealis. Cytoskeleton (Hoboken) 2018; 75:244-257. [PMID: 29573195 DOI: 10.1002/cm.21444] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 11/05/2022]
Abstract
Egg extracts of the African clawed frog Xenopus laevis have provided a cell-free system instrumental in elucidating events of the cell cycle, including mechanisms of spindle assembly. Comparison with extracts from the diploid Western clawed frog, Xenopus tropicalis, which is smaller at the organism, cellular and subcellular levels, has enabled the identification of spindle size scaling factors. We set out to characterize the Marsabit clawed frog, Xenopus borealis, which is intermediate in size between the two species, but more recently diverged in evolution from X. laevis than X. tropicalis. X. borealis eggs were slightly smaller than those of X. laevis, and slightly smaller spindles were assembled in egg extracts. Interestingly, microtubule distribution across the length of the X. borealis spindles differed from both X. laevis and X. tropicalis. Extract mixing experiments revealed common scaling phenomena among Xenopus species, while characterization of spindle factors katanin, TPX2, and Ran indicate that X. borealis spindles possess both X. laevis and X. tropicalis features. Thus, X. borealis egg extract provides a third in vitro system to investigate interspecies scaling and spindle morphometric variation.
Collapse
Affiliation(s)
- Maiko Kitaoka
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Rebecca Heald
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Romain Gibeaux
- Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| |
Collapse
|
59
|
Coming into Focus: Mechanisms of Microtubule Minus-End Organization. Trends Cell Biol 2018; 28:574-588. [PMID: 29571882 DOI: 10.1016/j.tcb.2018.02.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/17/2018] [Accepted: 02/27/2018] [Indexed: 11/22/2022]
Abstract
Microtubule organization has a crucial role in regulating cell architecture. The geometry of microtubule arrays strongly depends on the distribution of sites responsible for microtubule nucleation and minus-end attachment. In cycling animal cells, the centrosome often represents a dominant microtubule-organizing center (MTOC). However, even in cells with a radial microtubule system, many microtubules are not anchored at the centrosome, but are instead linked to the Golgi apparatus or other structures. Non-centrosomal microtubules predominate in many types of differentiated cell and in mitotic spindles. In this review, we discuss recent advances in understanding how the organization of centrosomal and non-centrosomal microtubule networks is controlled by proteins involved in microtubule nucleation and specific factors that recognize free microtubule minus ends and regulate their localization and dynamics.
Collapse
|
60
|
Microtubule-Organizing Centers: Towards a Minimal Parts List. Trends Cell Biol 2017; 28:176-187. [PMID: 29173799 DOI: 10.1016/j.tcb.2017.10.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/20/2017] [Accepted: 10/23/2017] [Indexed: 12/17/2022]
Abstract
Despite decades of molecular analysis of the centrosome, an important microtubule-organizing center (MTOC) of animal cells, the molecular basis of microtubule organization remains obscure. A major challenge is the sheer complexity of the interplay of the hundreds of proteins that constitute the centrosome. However, this complexity owes not only to the centrosome's role as a MTOC but also to the requirements of its duplication cycle and to various other functions such as the formation of cilia, the integration of various signaling pathways, and the organization of actin filaments. Thus, rather than using the parts lists to reconstruct the centrosome, we propose to identify the subset of proteins minimally needed to assemble a MTOC and to study this process at non-centrosomal sites.
Collapse
|
61
|
Zhang R, Roostalu J, Surrey T, Nogales E. Structural insight into TPX2-stimulated microtubule assembly. eLife 2017; 6. [PMID: 29120325 PMCID: PMC5679754 DOI: 10.7554/elife.30959] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/29/2017] [Indexed: 11/13/2022] Open
Abstract
During mitosis and meiosis, microtubule (MT) assembly is locally upregulated by the chromatin-dependent Ran-GTP pathway. One of its key targets is the MT-associated spindle assembly factor TPX2. The molecular mechanism of how TPX2 stimulates MT assembly remains unknown because structural information about the interaction of TPX2 with MTs is lacking. Here, we determine the cryo-electron microscopy structure of a central region of TPX2 bound to the MT surface. TPX2 uses two flexibly linked elements ('ridge' and 'wedge') in a novel interaction mode to simultaneously bind across longitudinal and lateral tubulin interfaces. These MT-interacting elements overlap with the binding site of importins on TPX2. Fluorescence microscopy-based in vitro reconstitution assays reveal that this interaction mode is critical for MT binding and facilitates MT nucleation. Together, our results suggest a molecular mechanism of how the Ran-GTP gradient can regulate TPX2-dependent MT formation.
Collapse
Affiliation(s)
- Rui Zhang
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, United States
| | | | | | - Eva Nogales
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, United States.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
62
|
Sanders AAWM, Chang K, Zhu X, Thoppil RJ, Holmes WR, Kaverina I. Nonrandom γ-TuNA-dependent spatial pattern of microtubule nucleation at the Golgi. Mol Biol Cell 2017; 28:3181-3192. [PMID: 28931596 PMCID: PMC5687021 DOI: 10.1091/mbc.e17-06-0425] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/05/2017] [Accepted: 09/13/2017] [Indexed: 01/12/2023] Open
Abstract
Noncentrosomal microtubule (MT) nucleation at the Golgi generates MT network asymmetry in motile vertebrate cells. Investigating the Golgi-derived MT (GDMT) distribution, we find that MT asymmetry arises from nonrandom nucleation sites at the Golgi (hotspots). Using computational simulations, we propose two plausible mechanistic models of GDMT nucleation leading to this phenotype. In the "cooperativity" model, formation of a single GDMT promotes further nucleation at the same site. In the "heterogeneous Golgi" model, MT nucleation is dramatically up-regulated at discrete and sparse locations within the Golgi. While MT clustering in hotspots is equally well described by both models, simulating MT length distributions within the cooperativity model fits the data better. Investigating the molecular mechanism underlying hotspot formation, we have found that hotspots are significantly smaller than a Golgi subdomain positive for scaffolding protein AKAP450, which is thought to recruit GDMT nucleation factors. We have further probed potential roles of known GDMT-promoting molecules, including γ-TuRC-mediated nucleation activator (γ-TuNA) domain-containing proteins and MT stabilizer CLASPs. While both γ-TuNA inhibition and lack of CLASPs resulted in drastically decreased GDMT nucleation, computational modeling revealed that only γ-TuNA inhibition suppressed hotspot formation. We conclude that hotspots require γ-TuNA activity, which facilitates clustered GDMT nucleation at distinct Golgi sites.
Collapse
Affiliation(s)
- Anna A W M Sanders
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Kevin Chang
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Xiaodong Zhu
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - Roslin J Thoppil
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| | - William R Holmes
- Physics and Astronomy, Vanderbilt University, Nashville, TN 37240
| | - Irina Kaverina
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240
| |
Collapse
|
63
|
Affiliation(s)
- Raymundo Alfaro-Aco
- a Department of Molecular Biology , Princeton University , Princeton , NJ , USA
| | - Sabine Petry
- a Department of Molecular Biology , Princeton University , Princeton , NJ , USA
| |
Collapse
|