1
|
Simoes-da-Silva MM, Barisic M. How does the tubulin code facilitate directed cell migration? Biochem Soc Trans 2025; 53:BST20240841. [PMID: 39998313 DOI: 10.1042/bst20240841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/03/2025] [Accepted: 02/07/2025] [Indexed: 02/26/2025]
Abstract
Besides being a component of the cytoskeleton that provides structural integrity to the cell, microtubules also serve as tracks for intracellular transport. As the building units of the mitotic spindle, microtubules distribute chromosomes during cell division. By distributing organelles, vesicles, and proteins, they play a pivotal role in diverse cellular processes, including cell migration, during which they reorganize to facilitate cell polarization. Structurally, microtubules are built up of α/β-tubulin dimers, which consist of various tubulin isotypes that undergo multiple post-translational modifications (PTMs). These PTMs allow microtubules to differentiate into functional subsets, influencing the associated processes. This text explores the current understanding of the roles of tubulin PTMs in cell migration, particularly detyrosination and acetylation, and their implications in human diseases.
Collapse
Affiliation(s)
| | - Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Institute, Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Kok M, Huber F, Kalisch SM, Dogterom M. EB3-informed dynamics of the microtubule stabilizing cap during stalled growth. Biophys J 2025; 124:227-244. [PMID: 39604262 PMCID: PMC11788501 DOI: 10.1016/j.bpj.2024.11.3314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/16/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
Microtubule stability is known to be governed by a stabilizing GTP/GDP-Pi cap, but the exact relation between growth velocity, GTP hydrolysis, and catastrophes remains unclear. We investigate the dynamics of the stabilizing cap through in vitro reconstitution of microtubule dynamics in contact with microfabricated barriers, using the plus-end binding protein GFP-EB3 as a marker for the nucleotide state of the tip. The interaction of growing microtubules with steric objects is known to slow down microtubule growth and accelerate catastrophes. We show that the lifetime distributions of stalled microtubules, as well as the corresponding lifetime distributions of freely growing microtubules, can be fully described with a simple phenomenological 1D model based on noisy microtubule growth and a single EB3-dependent hydrolysis rate. This same model is furthermore capable of explaining both the previously reported mild catastrophe dependence on microtubule growth rates and the catastrophe statistics during tubulin washout experiments.
Collapse
Affiliation(s)
- Maurits Kok
- Department of Bionanoscience, Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Florian Huber
- Department of Bionanoscience, Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands; Netherlands eScience Center, Amsterdam, the Netherlands; Center for Digitalisation and Digitality, Düsseldorf University of Applied Sciences, Düsseldorf, Germany
| | - Svenja-Marei Kalisch
- Department of Bionanoscience, Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands
| | - Marileen Dogterom
- Department of Bionanoscience, Kavli Institute of Nanoscience, Faculty of Applied Sciences, Delft University of Technology, Delft, the Netherlands.
| |
Collapse
|
3
|
Ju RJ, Falconer AD, Schmidt CJ, Enriquez Martinez MA, Dean KM, Fiolka RP, Sester DP, Nobis M, Timpson P, Lomakin AJ, Danuser G, White MD, Haass NK, Oelz DB, Stehbens SJ. Compression-dependent microtubule reinforcement enables cells to navigate confined environments. Nat Cell Biol 2024; 26:1520-1534. [PMID: 39160291 DOI: 10.1038/s41556-024-01476-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/11/2024] [Indexed: 08/21/2024]
Abstract
Cells migrating through complex three-dimensional environments experience considerable physical challenges, including tensile stress and compression. To move, cells need to resist these forces while also squeezing the large nucleus through confined spaces. This requires highly coordinated cortical contractility. Microtubules can both resist compressive forces and sequester key actomyosin regulators to ensure appropriate activation of contractile forces. Yet, how these two roles are integrated to achieve nuclear transmigration in three dimensions is largely unknown. Here, we demonstrate that compression triggers reinforcement of a dedicated microtubule structure at the rear of the nucleus by the mechanoresponsive recruitment of cytoplasmic linker-associated proteins, which dynamically strengthens and repairs the lattice. These reinforced microtubules form the mechanostat: an adaptive feedback mechanism that allows the cell to both withstand compressive force and spatiotemporally organize contractility signalling pathways. The microtubule mechanostat facilitates nuclear positioning and coordinates force production to enable the cell to pass through constrictions. Disruption of the mechanostat imbalances cortical contractility, stalling migration and ultimately resulting in catastrophic cell rupture. Our findings reveal a role for microtubules as cellular sensors that detect and respond to compressive forces, enabling movement and ensuring survival in mechanically demanding environments.
Collapse
Affiliation(s)
- Robert J Ju
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Alistair D Falconer
- School of Mathematics and Physics, University of Queensland, Brisbane, Queensland, Australia
| | - Christanny J Schmidt
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Marco A Enriquez Martinez
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia
| | - Kevin M Dean
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Reto P Fiolka
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David P Sester
- TRI Flow Cytometry Suite (TRI.fcs), Translational Research Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Max Nobis
- Faculty of Medicine, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Faculty of Medicine, St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Paul Timpson
- Faculty of Medicine, The Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Division, St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia
- Faculty of Medicine, St. Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Alexis J Lomakin
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
- Institute of Medical Chemistry and Pathobiochemistry, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Gaudenz Danuser
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Cecil H. and Ida Green Centre for Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Melanie D White
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Nikolas K Haass
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia
| | - Dietmar B Oelz
- School of Mathematics and Physics, University of Queensland, Brisbane, Queensland, Australia.
| | - Samantha J Stehbens
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland, Australia.
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia.
- Frazer Institute, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
4
|
Thery M, Akhmanova A. Confined migration: Microtubules control the cell rear. Curr Biol 2024; 34:R728-R731. [PMID: 39106829 DOI: 10.1016/j.cub.2024.06.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
Abstract
Cell migration through complex 3D environments relies on the interplay between actin and microtubules. A new study shows that, when cells pass through narrow constrictions, CLASP-dependent microtubule stabilisation at the cell rear controls actomyosin contractility to enable nuclear translocation and preserve cell integrity.
Collapse
Affiliation(s)
- Manuel Thery
- CytoMorpho Lab, LPCV, UMR5168, Université Grenoble-Alpes, CEA/INRA/CNRS, Interdisciplinary Research Institute of Grenoble, 17 rue des Martyrs, 38054 Grenoble, France; CytoMorpho Lab, CBI, UMR8132, Université Paris Sciences et Lettres, Ecole Supérieure de Physique et Chimie Industrielles de la Ville de Paris, CEA/CNRS, Institut Pierre Gilles De Gennes, 6 rue Jean Calvin, 75005 Paris, France.
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 Utrecht, The Netherlands.
| |
Collapse
|
5
|
Conboy JP, Istúriz Petitjean I, van der Net A, Koenderink GH. How cytoskeletal crosstalk makes cells move: Bridging cell-free and cell studies. BIOPHYSICS REVIEWS 2024; 5:021307. [PMID: 38840976 PMCID: PMC11151447 DOI: 10.1063/5.0198119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Cell migration is a fundamental process for life and is highly dependent on the dynamical and mechanical properties of the cytoskeleton. Intensive physical and biochemical crosstalk among actin, microtubules, and intermediate filaments ensures their coordination to facilitate and enable migration. In this review, we discuss the different mechanical aspects that govern cell migration and provide, for each mechanical aspect, a novel perspective by juxtaposing two complementary approaches to the biophysical study of cytoskeletal crosstalk: live-cell studies (often referred to as top-down studies) and cell-free studies (often referred to as bottom-up studies). We summarize the main findings from both experimental approaches, and we provide our perspective on bridging the two perspectives to address the open questions of how cytoskeletal crosstalk governs cell migration and makes cells move.
Collapse
Affiliation(s)
- James P. Conboy
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Irene Istúriz Petitjean
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Anouk van der Net
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Gijsje H. Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| |
Collapse
|
6
|
Schmidt CJ, Stehbens SJ. Microtubule control of migration: Coordination in confinement. Curr Opin Cell Biol 2024; 86:102289. [PMID: 38041936 DOI: 10.1016/j.ceb.2023.102289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 12/04/2023]
Abstract
The microtubule cytoskeleton has a well-established, instrumental role in coordinating cell migration. Decades of research has focused on understanding how microtubules couple intracellular trafficking with cortical targeting and spatial organization of signaling to facilitate locomotion. Movement in physically challenging environments requires coordination of forces generated by the actin cytoskeleton to drive cell shape changes, with microtubules acting to spatially regulate contractility. Recent work has demonstrated that the mechanical properties of microtubules are adaptive to stress, leading to a new understanding of their roles in cell migration. Herein we review new developments in how microtubules sense and adapt to changes in the physical properties of their environment during migration. We frame our discussion around our current understanding of how microtubules target cell-matrix adhesions, and their role in the spatiotemporal coordination of signaling to form mechano feedback loops. We expand on how these mechanisms may influence cell morphology in confined three-dimensional settings, and the importance of locally tuning the mechanical stability of polymers in response to mechanical cues. Finally, we discuss new roles for Golgi-derived microtubules in mechanosensing, and how preferential motor use may influence polymer stability to resist the physical constraints cells experience in confined environments.
Collapse
Affiliation(s)
- Christanny J Schmidt
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia
| | - Samantha J Stehbens
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, 4072, Australia; Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, Australia.
| |
Collapse
|
7
|
Gélin M, Schaeffer A, Gaillard J, Guérin C, Vianay B, Orhant-Prioux M, Braun M, Leterrier C, Blanchoin L, Théry M. Microtubules under mechanical pressure can breach dense actin networks. J Cell Sci 2023; 136:jcs261667. [PMID: 37870087 DOI: 10.1242/jcs.261667] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/24/2023] Open
Abstract
The crosstalk between the actin network and microtubules is essential for cell polarity. It orchestrates microtubule organization within the cell, driven by the asymmetry of actin architecture along the cell periphery. The physical intertwining of these networks regulates spatial organization and force distribution in the microtubule network. Although their biochemical interactions are becoming clearer, the mechanical aspects remain less understood. To explore this mechanical interplay, we developed an in vitro reconstitution assay to investigate how dynamic microtubules interact with various actin filament structures. Our findings revealed that microtubules can align and move along linear actin filament bundles through polymerization force. However, they are unable to pass through when encountering dense branched actin meshworks, similar to those present in the lamellipodium along the periphery of the cell. Interestingly, immobilizing microtubules through crosslinking with actin or other means allow the buildup of pressure, enabling them to breach these dense actin barriers. This mechanism offers insights into microtubule progression towards the cell periphery, with them overcoming obstacles within the denser parts of the actin network and ultimately contributing to cell polarity establishment.
Collapse
Affiliation(s)
- Matthieu Gélin
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Alexandre Schaeffer
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Jérémie Gaillard
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Christophe Guérin
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Benoit Vianay
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
| | - Magali Orhant-Prioux
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Marcus Braun
- Institute of Biotechnology, Czech Academy of Sciences, BIOCEV, 25250 Vestec, Prague West, Czech Republic
| | - Christophe Leterrier
- Aix Marseille Université, CNRS, INP UMR7051, NeuroCyto, 13385, Marseille, France
| | - Laurent Blanchoin
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| | - Manuel Théry
- Université Paris cité, CEA, INSERM, Institut de Recherche Saint Louis, UMR976 HIPI, CytoMorpho Lab, Avenue Claude Vellefaux, 75010 Paris, France
- Université Grenoble-Alpes, CEA, CNRS, INRA, Interdisciplinary Research Institute of Grenoble, UMR5168-LPCV, CytoMorpho Lab, Avenue des Martyrs, 38054 Grenoble, France
| |
Collapse
|
8
|
Lavrsen K, Rajendraprasad G, Leda M, Eibes S, Vitiello E, Katopodis V, Goryachev AB, Barisic M. Microtubule detyrosination drives symmetry breaking to polarize cells for directed cell migration. Proc Natl Acad Sci U S A 2023; 120:e2300322120. [PMID: 37216553 PMCID: PMC10235987 DOI: 10.1073/pnas.2300322120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
To initiate directed movement, cells must become polarized, establishing a protrusive leading edge and a contractile trailing edge. This symmetry-breaking process involves reorganization of cytoskeleton and asymmetric distribution of regulatory molecules. However, what triggers and maintains this asymmetry during cell migration remains largely elusive. Here, we established a micropatterning-based 1D motility assay to investigate the molecular basis of symmetry breaking required for directed cell migration. We show that microtubule (MT) detyrosination drives cell polarization by directing kinesin-1-based transport of the adenomatous polyposis coli (APC) protein to cortical sites. This is essential for the formation of cell's leading edge during 1D and 3D cell migration. These data, combined with biophysical modeling, unveil a key role for MT detyrosination in the generation of a positive feedback loop linking MT dynamics and kinesin-1-based transport. Thus, symmetry breaking during cell polarization relies on a feedback loop driven by MT detyrosination that supports directed cell migration.
Collapse
Affiliation(s)
- Kirstine Lavrsen
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center, 2100Copenhagen, Denmark
| | - Girish Rajendraprasad
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center, 2100Copenhagen, Denmark
| | - Marcin Leda
- Centre for Synthetic and Systems Biology, University of Edinburgh, EdinburghEH9 3BF, United Kingdom
| | - Susana Eibes
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center, 2100Copenhagen, Denmark
| | - Elisa Vitiello
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center, 2100Copenhagen, Denmark
| | - Vasileios Katopodis
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center, 2100Copenhagen, Denmark
| | - Andrew B. Goryachev
- Centre for Synthetic and Systems Biology, University of Edinburgh, EdinburghEH9 3BF, United Kingdom
| | - Marin Barisic
- Cell Division and Cytoskeleton, Danish Cancer Society Research Center, 2100Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200Copenhagen, Denmark
| |
Collapse
|
9
|
Hohmann T, Hohmann U, Dehghani F. MACC1-induced migration in tumors: Current state and perspective. Front Oncol 2023; 13:1165676. [PMID: 37051546 PMCID: PMC10084939 DOI: 10.3389/fonc.2023.1165676] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Abstract
Malignant tumors are still a global, heavy health burden. Many tumor types cannot be treated curatively, underlining the need for new treatment targets. In recent years, metastasis associated in colon cancer 1 (MACC1) was identified as a promising biomarker and drug target, as it is promoting tumor migration, initiation, proliferation, and others in a multitude of solid cancers. Here, we will summarize the current knowledge about MACC1-induced tumor cell migration with a special focus on the cytoskeletal and adhesive systems. In addition, a brief overview of several in vitro models used for the analysis of cell migration is given. In this context, we will point to issues with the currently most prevalent models used to study MACC1-dependent migration. Lastly, open questions about MACC1-dependent effects on tumor cell migration will be addressed.
Collapse
|
10
|
Wang W, Li W, Pan L, Li L, Xu Y, Wang Y, Zhang X, Zhang S. Dynamic Regulation Genes at Microtubule Plus Ends: A Novel Class of Glioma Biomarkers. BIOLOGY 2023; 12:biology12030488. [PMID: 36979179 PMCID: PMC10045452 DOI: 10.3390/biology12030488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023]
Abstract
Simple Summary Microtubule plus-end-related genes (MPERGs) encode a group of proteins that specifically aggregate at the microtubule plus ends to play critical biological roles in the cell cycle, cell movement, ciliogenesis, and neuronal development by coordinating microtubule assembly and dynamics; however, the MPERG correlations and their clinical significance in glioma are not fully understood. This study is the first to systematically analyze and define a seven-gene signature (CTTNBP2, KIF18A, NAV1, SLAIN2, SRCIN1, TRIO, and TTBK2) and nomogram model closely associated with clinical factors and the tumor microenvironment as a reliable and independent prognostic biomarker to guide personalized choices of immunotherapy and chemotherapy for glioma patients. Abstract Glioma is the most prevalent and aggressive primary nervous system tumor with an unfavorable prognosis. Microtubule plus-end-related genes (MPERGs) play critical biological roles in the cell cycle, cell movement, ciliogenesis, and neuronal development by coordinating microtubule assembly and dynamics. This research seeks to systematically explore the oncological characteristics of these genes in microtubule-enriched glioma, focusing on developing a novel MPERG-based prognostic signature to improve the prognosis and provide more treatment options for glioma patients. First, we thoroughly analyzed and identified 45 differentially expressed MPERGs in glioma. Based on these genes, glioma patients were well distinguished into two subgroups with survival and tumor microenvironment infiltration differences. Next, we further screened the independent prognostic genes (CTTNBP2, KIF18A, NAV1, SLAIN2, SRCIN1, TRIO, and TTBK2) using 36 prognostic-related differentially expressed MPERGs to construct a signature with risk stratification and prognostic prediction ability. An increased risk score was related to the malignant progression of glioma. Therefore, we also designed a nomogram model containing clinical factors to facilitate the clinical use of the risk signature. The prediction accuracy of the signature and nomogram model was verified using The Cancer Genome Atlas and Chinese Glioma Genome Atlas datasets. Finally, we examined the connection between the signature and tumor microenvironment. The signature positively correlated with tumor microenvironment infiltration, especially immunoinhibitors and the tumor mutation load, and negatively correlated with microsatellite instability and cancer stemness. More importantly, immune checkpoint blockade treatment and drug sensitivity analyses confirmed that this prognostic signature was helpful in anticipating the effect of immunotherapy and chemotherapy. In conclusion, this research is the first study to define and validate an MPERG-based signature closely associated with the tumor microenvironment as a reliable and independent prognostic biomarker to guide personalized choices of immunotherapy and chemotherapy for glioma patients.
Collapse
Affiliation(s)
- Wenwen Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Weilong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Lifang Pan
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310006, China
| | - Lingjie Li
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310006, China
| | - Yasi Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yuqing Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaochen Zhang
- Department of Medical Oncology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310006, China
- Correspondence: (X.Z.); (S.Z.); Tel./Fax: +86-571-5600-7650 (S.Z.)
| | - Shirong Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Affiliated Hangzhou First People’s Hospital, Hangzhou 310053, China
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
- Correspondence: (X.Z.); (S.Z.); Tel./Fax: +86-571-5600-7650 (S.Z.)
| |
Collapse
|
11
|
Legátová A, Pelantová M, Rösel D, Brábek J, Škarková A. The emerging role of microtubules in invasion plasticity. Front Oncol 2023; 13:1118171. [PMID: 36860323 PMCID: PMC9969133 DOI: 10.3389/fonc.2023.1118171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
The ability of cells to switch between different invasive modes during metastasis, also known as invasion plasticity, is an important characteristic of tumor cells that makes them able to resist treatment targeted to a particular invasion mode. Due to the rapid changes in cell morphology during the transition between mesenchymal and amoeboid invasion, it is evident that this process requires remodeling of the cytoskeleton. Although the role of the actin cytoskeleton in cell invasion and plasticity is already quite well described, the contribution of microtubules is not yet fully clarified. It is not easy to infer whether destabilization of microtubules leads to higher invasiveness or the opposite since the complex microtubular network acts differently in diverse invasive modes. While mesenchymal migration typically requires microtubules at the leading edge of migrating cells to stabilize protrusions and form adhesive structures, amoeboid invasion is possible even in the absence of long, stable microtubules, albeit there are also cases of amoeboid cells where microtubules contribute to effective migration. Moreover, complex crosstalk of microtubules with other cytoskeletal networks participates in invasion regulation. Altogether, microtubules play an important role in tumor cell plasticity and can be therefore targeted to affect not only cell proliferation but also invasive properties of migrating cells.
Collapse
Affiliation(s)
- Anna Legátová
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Markéta Pelantová
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Jan Brábek
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia
| | - Aneta Škarková
- Department of Cell Biology, Charles University, Prague, Czechia,Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Vestec u Prahy, Czechia,*Correspondence: Aneta Škarková,
| |
Collapse
|
12
|
van den Berg CM, Volkov VA, Schnorrenberg S, Huang Z, Stecker KE, Grigoriev I, Gilani S, Frikstad KAM, Patzke S, Zimmermann T, Dogterom M, Akhmanova A. CSPP1 stabilizes growing microtubule ends and damaged lattices from the luminal side. J Cell Biol 2023; 222:213861. [PMID: 36752787 PMCID: PMC9948759 DOI: 10.1083/jcb.202208062] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/14/2022] [Accepted: 01/13/2023] [Indexed: 02/09/2023] Open
Abstract
Microtubules are dynamic cytoskeletal polymers, and their organization and stability are tightly regulated by numerous cellular factors. While regulatory proteins controlling the formation of interphase microtubule arrays and mitotic spindles have been extensively studied, the biochemical mechanisms responsible for generating stable microtubule cores of centrioles and cilia are poorly understood. Here, we used in vitro reconstitution assays to investigate microtubule-stabilizing properties of CSPP1, a centrosome and cilia-associated protein mutated in the neurodevelopmental ciliopathy Joubert syndrome. We found that CSPP1 preferentially binds to polymerizing microtubule ends that grow slowly or undergo growth perturbations and, in this way, resembles microtubule-stabilizing compounds such as taxanes. Fluorescence microscopy and cryo-electron tomography showed that CSPP1 is deposited in the microtubule lumen and inhibits microtubule growth and shortening through two separate domains. CSPP1 also specifically recognizes and stabilizes damaged microtubule lattices. These data help to explain how CSPP1 regulates the elongation and stability of ciliary axonemes and other microtubule-based structures.
Collapse
Affiliation(s)
- Cyntha M. van den Berg
- https://ror.org/04pp8hn57Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Vladimir A. Volkov
- https://ror.org/04pp8hn57Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands,https://ror.org/02e2c7k09Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, The Netherlands
| | | | - Ziqiang Huang
- EMBL Imaging Centre, EMBL-Heidelberg, Heidelberg, Germany
| | - Kelly E. Stecker
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands,Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Ilya Grigoriev
- https://ror.org/04pp8hn57Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Sania Gilani
- https://ror.org/00j9c2840Department of Radiation Biology, Institute of Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway,Department of Molecular Cell Biology, Institute of Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Kari-Anne M. Frikstad
- https://ror.org/00j9c2840Department of Radiation Biology, Institute of Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Sebastian Patzke
- https://ror.org/00j9c2840Department of Radiation Biology, Institute of Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | | | - Marileen Dogterom
- https://ror.org/02e2c7k09Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, Delft, The Netherlands
| | - Anna Akhmanova
- https://ror.org/04pp8hn57Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands,Correspondence to Anna Akhmanova:
| |
Collapse
|
13
|
Luo W, Demidov V, Shen Q, Girão H, Chakraborty M, Maiorov A, Ataullakhanov FI, Lin C, Maiato H, Grishchuk EL. CLASP2 recognizes tubulins exposed at the microtubule plus-end in a nucleotide state-sensitive manner. SCIENCE ADVANCES 2023; 9:eabq5404. [PMID: 36598991 PMCID: PMC9812398 DOI: 10.1126/sciadv.abq5404] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 11/23/2022] [Indexed: 05/28/2023]
Abstract
CLASPs (cytoplasmic linker-associated proteins) are ubiquitous stabilizers of microtubule dynamics, but their molecular targets at the microtubule plus-end are not understood. Using DNA origami-based reconstructions, we show that clusters of human CLASP2 form a load-bearing bond with terminal non-GTP tubulins at the stabilized microtubule tip. This activity relies on the unconventional TOG2 domain of CLASP2, which releases its high-affinity bond with non-GTP dimers upon their conversion into polymerization-competent GTP-tubulins. The ability of CLASP2 to recognize nucleotide-specific tubulin conformation and stabilize the catastrophe-promoting non-GTP tubulins intertwines with the previously underappreciated exchange between GDP and GTP at terminal tubulins. We propose that TOG2-dependent stabilization of sporadically occurring non-GTP tubulins represents a distinct molecular mechanism to suppress catastrophe at the freely assembling microtubule ends and to promote persistent tubulin assembly at the load-bearing tethered ends, such as at the kinetochores in dividing cells.
Collapse
Affiliation(s)
- Wangxi Luo
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Vladimir Demidov
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qi Shen
- Department of Cell Biology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
| | - Hugo Girão
- Chromosome Instability & Dynamics Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Manas Chakraborty
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aleksandr Maiorov
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fazly I. Ataullakhanov
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, 119991 Moscow, Russian Federation
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region 141701, Russian Federation
| | - Chenxiang Lin
- Department of Cell Biology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
- Nanobiology Institute, Yale University, West Haven, CT 06516, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Helder Maiato
- Chromosome Instability & Dynamics Group, Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Cell Division Group, Department of Biomedicine, Faculdade de Medicina, Universidade do Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ekaterina L. Grishchuk
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
14
|
Merino-Casallo F, Gomez-Benito MJ, Hervas-Raluy S, Garcia-Aznar JM. Unravelling cell migration: defining movement from the cell surface. Cell Adh Migr 2022; 16:25-64. [PMID: 35499121 PMCID: PMC9067518 DOI: 10.1080/19336918.2022.2055520] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Cell motility is essential for life and development. Unfortunately, cell migration is also linked to several pathological processes, such as cancer metastasis. Cells' ability to migrate relies on many actors. Cells change their migratory strategy based on their phenotype and the properties of the surrounding microenvironment. Cell migration is, therefore, an extremely complex phenomenon. Researchers have investigated cell motility for more than a century. Recent discoveries have uncovered some of the mysteries associated with the mechanisms involved in cell migration, such as intracellular signaling and cell mechanics. These findings involve different players, including transmembrane receptors, adhesive complexes, cytoskeletal components , the nucleus, and the extracellular matrix. This review aims to give a global overview of our current understanding of cell migration.
Collapse
Affiliation(s)
- Francisco Merino-Casallo
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Maria Jose Gomez-Benito
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Silvia Hervas-Raluy
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Jose Manuel Garcia-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
15
|
Meiring JCM, Grigoriev I, Nijenhuis W, Kapitein LC, Akhmanova A. Opto-katanin, an optogenetic tool for localized, microtubule disassembly. Curr Biol 2022; 32:4660-4674.e6. [PMID: 36174574 DOI: 10.1016/j.cub.2022.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/01/2022] [Accepted: 09/06/2022] [Indexed: 11/30/2022]
Abstract
Microtubules are cytoskeletal polymers that separate chromosomes during mitosis and serve as rails for intracellular transport and organelle positioning. Manipulation of microtubules is widely used in cell and developmental biology, but tools for precise subcellular spatiotemporal control of microtubules are currently lacking. Here, we describe a light-activated system for localized recruitment of the microtubule-severing enzyme katanin. This system, named opto-katanin, uses targeted illumination with blue light to induce rapid, localized, and reversible microtubule depolymerization. This tool allows precise clearing of a subcellular region of microtubules while preserving the rest of the microtubule network, demonstrating that regulation of katanin recruitment to microtubules is sufficient to control its severing activity. The tool is not toxic in the absence of blue light and can be used to disassemble both dynamic and stable microtubules in primary neurons as well as in dividing cells. We show that opto-katanin can be used to locally block vesicle transport and to clarify the dependence of organelle morphology and dynamics on microtubules. Specifically, our data indicate that microtubules are not required for the maintenance of the Golgi stacks or the tubules of the endoplasmic reticulum but are needed for the formation of new membrane tubules. Finally, we demonstrate that this tool can be applied to study the contribution of microtubules to cell mechanics by showing that microtubule bundles can exert forces constricting the nucleus.
Collapse
Affiliation(s)
- Joyce C M Meiring
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands
| | - Ilya Grigoriev
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands
| | - Wilco Nijenhuis
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands; Center for Living Technologies, Eindhoven-Wageningen-Utrecht Alliance, UMC Utrecht, Utrecht 3584 CB, the Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands; Center for Living Technologies, Eindhoven-Wageningen-Utrecht Alliance, UMC Utrecht, Utrecht 3584 CB, the Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan, Utrecht 3584 CS, the Netherlands.
| |
Collapse
|
16
|
MALAT1 in colorectal cancer: Its implication as a diagnostic, prognostic, and predictive biomarker. Gene 2022; 843:146791. [PMID: 35961438 DOI: 10.1016/j.gene.2022.146791] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/26/2022] [Accepted: 08/05/2022] [Indexed: 12/13/2022]
Abstract
Metastasis Associated Lung Adenocarcinoma Transcript 1 (MALAT1), originally described as a prognostic biomarker remarkably linked with metastasis potential in lung cancer, has been identified as contributing to many diseases, including colorectal cancer (CRC). This long non-coding RNA (lncRNA) has come to the forefront of lncRNA research for its implications in cancer-related processes, such as cell proliferation and migration. In general, lncRNAs are recognized as enhancers, scaffolds, or decoys for a variety of oncogenes and tumor suppressors, although our understanding of lncRNA functions and mechanisms of action is still limited. Nowadays, cancer research is attracted to lncRNAs' ability to improve the early diagnosis of cancer, determine patients' prognosis, or predict therapy outcomes. In this review, we aimed to evaluate recent publications trying to uncover the cellular mechanisms of MALAT1-mediated regulation, and its potential exploitation in the management of CRC. The conclusions of this review provide robust support for the essential role of MALAT1 in CRC development and future personalized therapy.
Collapse
|
17
|
Pudelko K, Wieland A, Hennecke M, Räschle M, Bastians H. Increased Microtubule Growth Triggered by Microvesicle-mediated Paracrine Signaling is Required for Melanoma Cancer Cell Invasion. CANCER RESEARCH COMMUNICATIONS 2022; 2:366-379. [PMID: 36875714 PMCID: PMC9981201 DOI: 10.1158/2767-9764.crc-22-0010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/25/2022] [Accepted: 05/03/2022] [Indexed: 11/16/2022]
Abstract
The acquisition of cell invasiveness is the key transition from benign melanocyte hyperplasia to aggressive melanoma. Recent work has provided an intriguing new link between the presence of supernumerary centrosomes and increased cell invasion. Moreover, supernumerary centrosomes were shown to drive non-cell-autonomous invasion of cancer cells. Although centrosomes are the principal microtubule organizing centers, the role of dynamic microtubules for non-cell-autonomous invasion remains unexplored, in particular, in melanoma. We investigated the role of supernumerary centrosomes and dynamic microtubules in melanoma cell invasion and found that highly invasive melanoma cells are characterized by the presence of supernumerary centrosomes and by increased microtubule growth rates, both of which are functionally interlinked. We demonstrate that enhanced microtubule growth is required for increased three-dimensional melanoma cell invasion. Moreover, we show that the activity to enhance microtubule growth can be transferred onto adjacent noninvasive cells through microvesicles involving HER2. Hence, our study suggests that suppressing microtubule growth, either directly using anti-microtubule drugs or through HER2 inhibitors might be therapeutically beneficial to inhibit cell invasiveness and thus, metastasis of malignant melanoma. Significance This study shows that increased microtubule growth is required for melanoma cell invasion and can be transferred onto adjacent cells in a non-cell-autonomous manner through microvesicles involving HER2.
Collapse
Affiliation(s)
- Karoline Pudelko
- Institute of Molecular Oncology, Section for Cellular Oncology, Georg-August University Göttingen, University Medical Center Göttingen (UMG) and Göttingen Center for Molecular Biosciences (GZMB), Göttingen, Germany
| | - Angela Wieland
- Department of Molecular Genetics, Technical University of Kaiserslautern, Kaiserslautern, Germany
| | - Magdalena Hennecke
- Institute of Molecular Oncology, Section for Cellular Oncology, Georg-August University Göttingen, University Medical Center Göttingen (UMG) and Göttingen Center for Molecular Biosciences (GZMB), Göttingen, Germany
| | - Markus Räschle
- Department of Molecular Genetics, Technical University of Kaiserslautern, Kaiserslautern, Germany
| | - Holger Bastians
- Institute of Molecular Oncology, Section for Cellular Oncology, Georg-August University Göttingen, University Medical Center Göttingen (UMG) and Göttingen Center for Molecular Biosciences (GZMB), Göttingen, Germany
| |
Collapse
|
18
|
Akhmanova A, Kapitein LC. Mechanisms of microtubule organization in differentiated animal cells. Nat Rev Mol Cell Biol 2022; 23:541-558. [PMID: 35383336 DOI: 10.1038/s41580-022-00473-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Microtubules are polarized cytoskeletal filaments that serve as tracks for intracellular transport and form a scaffold that positions organelles and other cellular components and modulates cell shape and mechanics. In animal cells, the geometry, density and directionality of microtubule networks are major determinants of cellular architecture, polarity and proliferation. In dividing cells, microtubules form bipolar spindles that pull chromosomes apart, whereas in interphase cells, microtubules are organized in a cell type-specific fashion, which strongly correlates with cell physiology. In motile cells, such as fibroblasts and immune cells, microtubules are organized as radial asters, whereas in immotile epithelial and neuronal cells and in muscles, microtubules form parallel or antiparallel arrays and cortical meshworks. Here, we review recent work addressing how the formation of such microtubule networks is driven by the plethora of microtubule regulatory proteins. These include proteins that nucleate or anchor microtubule ends at different cellular structures and those that sever or move microtubules, as well as regulators of microtubule elongation, stability, bundling or modifications. The emerging picture, although still very incomplete, shows a remarkable diversity of cell-specific mechanisms that employ conserved building blocks to adjust microtubule organization in order to facilitate different cellular functions.
Collapse
Affiliation(s)
- Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
19
|
Mathias TJ, Ju JA, Lee RM, Thompson KN, Mull ML, Annis DA, Chang KT, Ory EC, Stemberger MB, Hotta T, Ohi R, Vitolo MI, Moutin MJ, Martin SS. Tubulin Carboxypeptidase Activity Promotes Focal Gelatin Degradation in Breast Tumor Cells and Induces Apoptosis in Breast Epithelial Cells That Is Overcome by Oncogenic Signaling. Cancers (Basel) 2022; 14:1707. [PMID: 35406479 PMCID: PMC8996877 DOI: 10.3390/cancers14071707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 01/27/2023] Open
Abstract
Post-translational modifications (PTMs) of the microtubule network impart differential functions across normal cell types and their cancerous counterparts. The removal of the C-terminal tyrosine of α-tubulin (deTyr-Tub) as performed by the tubulin carboxypeptidase (TCP) is of particular interest in breast epithelial and breast cancer cells. The recent discovery of the genetic identity of the TCP to be a vasohibin (VASH1/2) coupled with a small vasohibin-binding protein (SVBP) allows for the functional effect of this tubulin PTM to be directly tested for the first time. Our studies revealed the immortalized breast epithelial cell line MCF10A undergoes apoptosis following transfection with TCP constructs, but the addition of oncogenic KRas or Bcl-2/Bcl-xL overexpression prevents subsequent apoptotic induction in the MCF10A background. Functionally, an increase in deTyr-Tub via TCP transfection in MDA-MB-231 and Hs578t breast cancer cells leads to enhanced focal gelatin degradation. Given the elevated deTyr-Tub at invasive tumor fronts and the correlation with poor breast cancer survival, these new discoveries help clarify how the TCP synergizes with oncogene activation, increases focal gelatin degradation, and may correspond to increased tumor cell invasion. These connections could inform more specific microtubule-directed therapies to target deTyr-tubulin.
Collapse
Affiliation(s)
- Trevor J. Mathias
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Graduate Program in Molecular Medicine, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD 21201, USA
- Medical Scientist Training Program (MSTP), University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD 21201, USA
| | - Julia A. Ju
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Rachel M. Lee
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Keyata N. Thompson
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Makenzy L. Mull
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Graduate Program in Molecular Medicine, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD 21201, USA
| | - David A. Annis
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Graduate Program in Epidemiology and Human Genetics, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD 21201, USA
| | - Katarina T. Chang
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Graduate Program in Molecular Medicine, University of Maryland School of Medicine, 800 W. Baltimore St., Baltimore, MD 21201, USA
| | - Eleanor C. Ory
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Megan B. Stemberger
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Graduate Program in Biochemistry & Molecular Biology, University of Maryland School of Medicine, 108 N. Greene St., Baltimore, MD 21201, USA
| | - Takashi Hotta
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA; (T.H.); (R.O.)
| | - Ryoma Ohi
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA; (T.H.); (R.O.)
| | - Michele I. Vitolo
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | - Marie-Jo Moutin
- Grenoble Institut Neurosciences, University Grenoble Alpes, Inserm, U1216, CEA, CNRS, 38000 Grenoble, France;
| | - Stuart S. Martin
- Marlene and Stewart Greenebaum NCI Comprehensive Cancer Center, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA; (T.J.M.); (J.A.J.); (R.M.L.); (K.N.T.); (M.L.M.); (D.A.A.); (K.T.C.); (E.C.O.); (M.B.S.); (M.I.V.)
- Department of Pharmacology and Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA
- United States Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD 21201, USA
| |
Collapse
|
20
|
Lee G, Leech G, Lwin P, Michel J, Currie C, Rust MJ, Ross JL, McGorty RJ, Das M, Robertson-Anderson RM. Active cytoskeletal composites display emergent tunable contractility and restructuring. SOFT MATTER 2021; 17:10765-10776. [PMID: 34792082 PMCID: PMC9239752 DOI: 10.1039/d1sm01083b] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The cytoskeleton is a model active matter system that controls processes as diverse as cell motility and mechanosensing. While both active actomyosin dynamics and actin-microtubule interactions are key to the cytoskeleton's versatility and adaptability, an understanding of their interplay is lacking. Here, we couple microscale experiments with mechanistic modeling to elucidate how connectivity, rigidity, and force-generation affect emergent material properties in composite networks of actin, tubulin, and myosin. We use multi-spectral imaging, time-resolved differential dynamic microscopy and spatial image autocorrelation to show that ballistic contraction occurs in composites with sufficient flexibility and motor density, but that a critical fraction of microtubules is necessary to sustain controlled dynamics. The active double-network models we develop, which recapitulate our experimental findings, reveal that while percolated actomyosin networks are essential for contraction, only composites with comparable actin and microtubule densities can simultaneously resist mechanical stresses while supporting substantial restructuring. The comprehensive phase map we present not only provides important insight into the different routes the cytoskeleton can use to alter its dynamics and structure, but also serves as a much-needed blueprint for designing cytoskeleton-inspired materials that couple tunability with resilience and adaptability for diverse applications ranging from wound healing to soft robotics.
Collapse
Affiliation(s)
- Gloria Lee
- Department of Physics and Biophysics, University of San Diego, USA.
| | - Gregor Leech
- Department of Physics and Biophysics, University of San Diego, USA.
| | - Pancy Lwin
- School of Physics and Astronomy, Rochester Institute of Technology, USA
| | - Jonathan Michel
- School of Physics and Astronomy, Rochester Institute of Technology, USA
| | | | - Michael J Rust
- Department of Molecular Genetics and Cell Biology, University of Chicago, USA
| | | | - Ryan J McGorty
- Department of Physics and Biophysics, University of San Diego, USA.
| | - Moumita Das
- School of Physics and Astronomy, Rochester Institute of Technology, USA
| | | |
Collapse
|
21
|
Jang SD, Song J, Kim HA, Im CN, Khawar IA, Park JK, Kuh HJ. Anti-Cancer Activity Profiling of Chemotherapeutic Agents in 3D Co-Cultures of Pancreatic Tumor Spheroids with Cancer-Associated Fibroblasts and Macrophages. Cancers (Basel) 2021; 13:5955. [PMID: 34885065 PMCID: PMC8656537 DOI: 10.3390/cancers13235955] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 12/29/2022] Open
Abstract
Activated pancreatic stellate cells (aPSCs) and M2 macrophages modulate tumor progression and therapeutic efficacy in pancreatic ductal adenocarcinoma (PDAC) via epithelial-mesenchymal transition (EMT). Here, our aim was to analyze the anti-invasion effects of anti-cancer agents where EMT-inducing cancer-stroma interaction occurs under three-dimensional (3D) culture conditions. We used microfluidic channel chips to co-culture pancreatic tumor spheroids (TSs) with aPSCs and THP-1-derived M2 macrophages (M2 THP-1 cells) embedded in type I collagen. Under stromal cell co-culture conditions, PANC-1 TSs displayed elevated expression of EMT-related proteins and increased invasion and migration. When PANC-1 TSs were exposed to gemcitabine, 5-fluorouracil, oxaliplatin, or paclitaxel, 30-50% cells were found unaffected, with no significant changes in the dose-response profiles under stromal cell co-culture conditions. This indicated intrinsic resistance to these drugs and no further induction of drug resistance by stromal cells. Paclitaxel had a significant anti-invasion effect; in contrast, oxaliplatin did not show such effect despite its specific cytotoxicity in M2 THP-1 cells. Overall, our findings demonstrate that the TS-stroma co-culture model of PDAC is useful for activity profiling of anti-cancer agents against cancer and stromal cells, and analyzing the relationship between anti-stromal activity and anti-invasion effects.
Collapse
Affiliation(s)
- So-Dam Jang
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Jeeyeun Song
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Hyun-Ah Kim
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
| | - Chang-Nim Im
- Graduate Program for Future Medical Research Leaders, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Iftikhar Ali Khawar
- Graduate Program for Future Medical Research Leaders, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jong Kook Park
- Department of Biomedical Science, Research Institute for Bioscience & Biotechnology, Hallym University, Chuncheon 24252, Korea
| | - Hyo-Jeong Kuh
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul 06591, Korea
- Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
22
|
Zhovmer AS, Manning A, Smith C, Hayes JB, Burnette DT, Wang J, Cartagena-Rivera AX, Dokholyan NV, Singh RK, Tabdanov ED. Mechanical Counterbalance of Kinesin and Dynein Motors in a Microtubular Network Regulates Cell Mechanics, 3D Architecture, and Mechanosensing. ACS NANO 2021; 15:17528-17548. [PMID: 34677937 PMCID: PMC9291236 DOI: 10.1021/acsnano.1c04435] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Microtubules (MTs) and MT motor proteins form active 3D networks made of unstretchable cables with rod-like bending mechanics that provide cells with a dynamically changing structural scaffold. In this study, we report an antagonistic mechanical balance within the dynein-kinesin microtubular motor system. Dynein activity drives the microtubular network inward compaction, while isolated activity of kinesins bundles and expands MTs into giant circular bands that deform the cell cortex into discoids. Furthermore, we show that dyneins recruit MTs to sites of cell adhesion, increasing the topographic contact guidance of cells, while kinesins antagonize it via retraction of MTs from sites of cell adhesion. Actin-to-microtubule translocation of septin-9 enhances kinesin-MT interactions, outbalances the activity of kinesins over that of dyneins, and induces the discoid architecture of cells. These orthogonal mechanisms of MT network reorganization highlight the existence of an intricate mechanical balance between motor activities of kinesins and dyneins that controls cell 3D architecture, mechanics, and cell-microenvironment interactions.
Collapse
Affiliation(s)
- Alexander S. Zhovmer
- Center
for Biologics Evaluation and Research, U.S.
Food and Drug Administration, Silver Spring, Maryland 20903, United States
| | - Alexis Manning
- Center
for Biologics Evaluation and Research, U.S.
Food and Drug Administration, Silver Spring, Maryland 20903, United States
| | - Chynna Smith
- Section
on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - James B. Hayes
- Department
of Cell and Developmental Biology, Vanderbilt Medical Center, University of Vanderbilt, Nashville, Tennessee 37232, United States
| | - Dylan T. Burnette
- Department
of Cell and Developmental Biology, Vanderbilt Medical Center, University of Vanderbilt, Nashville, Tennessee 37232, United States
| | - Jian Wang
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
| | - Alexander X. Cartagena-Rivera
- Section
on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Nikolay V. Dokholyan
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
- Department
of Biochemistry & Molecular Biology, Penn State College of Medicine, Pennsylvania State University, Hershey, Pennsylvania 17033, United States
| | - Rakesh K. Singh
- Department
of Obstetrics and Gynecology, University
of Rochester Medical Center, Rochester, New York 14620, United States
| | - Erdem D. Tabdanov
- Department
of Pharmacology, Penn State College of Medicine, Pennsylvania State University, Hummelstown, Pennsylvania 17036, United States
| |
Collapse
|
23
|
Panzade S, Matis M. The Microtubule Minus-End Binding Protein Patronin Is Required for the Epithelial Remodeling in the Drosophila Abdomen. Front Cell Dev Biol 2021; 9:682083. [PMID: 34368132 PMCID: PMC8335404 DOI: 10.3389/fcell.2021.682083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/24/2021] [Indexed: 11/29/2022] Open
Abstract
In the developing Drosophila abdomen, the epithelial tissue displays extensive cytoskeletal remodeling. In stark contrast to the spatio-temporal control of the actin cytoskeleton, the regulation of microtubule architecture during epithelial morphogenesis has remained opaque. In particular, its role in cell motility remains unclear. Here, we show that minus-end binding protein Patronin is required for organizing microtubule arrays in histoblast cells that form the Drosophila abdomen. Loss of Patronin results in a dorsal cleft, indicating the compromised function of histoblasts. We further show that Patronin is polarized in these cells and is required for the formation of highly dynamic non-centrosomal microtubules in the migrating histoblasts. Thus, our study demonstrates that regulation of microtubule cytoskeleton through Patronin mediates epithelium remodeling.
Collapse
Affiliation(s)
- Sadhana Panzade
- Interfaculty Centre 'Cells in Motion,' University of Münster, Münster, Germany.,Institute of Cell Biology, Medical Faculty, University of Münster, Münster, Germany
| | - Maja Matis
- Interfaculty Centre 'Cells in Motion,' University of Münster, Münster, Germany.,Institute of Cell Biology, Medical Faculty, University of Münster, Münster, Germany
| |
Collapse
|
24
|
Torrino S, Grasset EM, Audebert S, Belhadj I, Lacoux C, Haynes M, Pisano S, Abélanet S, Brau F, Chan SY, Mari B, Oldham WM, Ewald AJ, Bertero T. Mechano-induced cell metabolism promotes microtubule glutamylation to force metastasis. Cell Metab 2021; 33:1342-1357.e10. [PMID: 34102109 DOI: 10.1016/j.cmet.2021.05.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/31/2021] [Accepted: 05/07/2021] [Indexed: 01/11/2023]
Abstract
Mechanical signals from the tumor microenvironment modulate cell mechanics and influence cell metabolism to promote cancer aggressiveness. Cells withstand external forces by adjusting the stiffness of their cytoskeleton. Microtubules (MTs) act as compression-bearing elements. Yet how cancer cells regulate MT dynamic in response to the locally constrained environment has remained unclear. Using breast cancer as a model of a disease in which mechanical signaling promotes disease progression, we show that matrix stiffening rewires glutamine metabolism to promote MT glutamylation and force MT stabilization, thereby promoting cell invasion. Pharmacologic inhibition of glutamine metabolism decreased MT glutamylation and affected their mechanical stabilization. Similarly, decreased MT glutamylation by overexpressing tubulin mutants lacking glutamylation site(s) decreased MT stability, thereby hampering cancer aggressiveness in vitro and in vivo. Together, our results decipher part of the enigmatic tubulin code that coordinates the fine-tunable properties of MT and link cell metabolism to MT dynamics and cancer aggressiveness.
Collapse
Affiliation(s)
| | - Eloise M Grasset
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stephane Audebert
- Aix-Marseille Univ, INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Ilyes Belhadj
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | | | - Meagan Haynes
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sabrina Pisano
- Université Côte d'Azur, CNRS, INSERM, IRCAN, Nice, France
| | | | - Frederic Brau
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | - Stephen Y Chan
- Center for Pulmonary Vascular Biology and Medicine, Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Bernard Mari
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France
| | - William M Oldham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew J Ewald
- Department of Cell Biology, Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
25
|
Zhou X, Lu J, Zhang Y, Guo J, Lin W, Van Norman JM, Qin Y, Zhu X, Yang Z. Membrane receptor-mediated mechano-transduction maintains cell integrity during pollen tube growth within the pistil. Dev Cell 2021; 56:1030-1042.e6. [PMID: 33756107 DOI: 10.1016/j.devcel.2021.02.030] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/22/2021] [Accepted: 02/26/2021] [Indexed: 12/11/2022]
Abstract
Invasive or penetrative growth is critical for developmental and reproductive processes (e.g., pollen tube penetration of pistils) and disease progression (e.g., cancer metastasis and fungal hyphae invasion). The invading or penetrating cells experience drastic changes in mechanical pressure from the surroundings and must balance growth with cell integrity. Here, we show that Arabidopsis pollen tubes sense and/or respond to mechanical changes via a cell-surface receptor kinase Buddha's Paper Seal 1 (BUPS1) while emerging from compressing female tissues. BUPS1-defective pollen tubes fail to maintain cell integrity after emergence from these tissues. The mechano-transduction function of BUPS1 is established by using a microfluidic channel device mimicking the mechanical features of the in vivo growth path. BUPS1-based mechano-transduction activates Rho-like GTPase from Plant 1 (ROP1) GTPase to promote exocytosis that facilitates secretion of BUPS1's ligands for mechanical signal amplification and cell wall rigidification in pollen tubes. These findings uncover a membrane receptor-based mechano-transduction system for cells to cope with the physical challenges during invasive or penetrative growth.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Botany and Plant Sciences and Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Jun Lu
- Shanghai Center for Plant Stress Biology, Chinese Academy of Sciences, Shanghai, China; National Key Laboratory of Plant Molecular Genetics, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Shanghai 200032, People's Republic of China
| | - Yuqin Zhang
- FAFU-UCR Joint Center for Horticultural Biology and Metabolomics, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Jingzhe Guo
- Department of Botany and Plant Sciences and Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Wenwei Lin
- Department of Botany and Plant Sciences and Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA; FAFU-UCR Joint Center for Horticultural Biology and Metabolomics, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Jaimie M Van Norman
- Department of Botany and Plant Sciences and Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA
| | - Yuan Qin
- Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Center for Genomics and Biotechnology, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Xiaoyue Zhu
- FAFU-UCR Joint Center for Horticultural Biology and Metabolomics, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Zhenbiao Yang
- Department of Botany and Plant Sciences and Institute for Integrative Genome Biology, University of California, Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
26
|
Hooikaas PJ, Damstra HG, Gros OJ, van Riel WE, Martin M, Smits YT, van Loosdregt J, Kapitein LC, Berger F, Akhmanova A. Kinesin-4 KIF21B limits microtubule growth to allow rapid centrosome polarization in T cells. eLife 2020; 9:62876. [PMID: 33346730 PMCID: PMC7817182 DOI: 10.7554/elife.62876] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/20/2020] [Indexed: 12/11/2022] Open
Abstract
When a T cell and an antigen-presenting cell form an immunological synapse, rapid dynein-driven translocation of the centrosome toward the contact site leads to reorganization of microtubules and associated organelles. Currently, little is known about how the regulation of microtubule dynamics contributes to this process. Here, we show that the knockout of KIF21B, a kinesin-4 linked to autoimmune disorders, causes microtubule overgrowth and perturbs centrosome translocation. KIF21B restricts microtubule length by inducing microtubule pausing typically followed by catastrophe. Catastrophe induction with vinblastine prevented microtubule overgrowth and was sufficient to rescue centrosome polarization in KIF21B-knockout cells. Biophysical simulations showed that a relatively small number of KIF21B molecules can restrict mirotubule length and promote an imbalance of dynein-mediated pulling forces that allows the centrosome to translocate past the nucleus. We conclude that proper control of microtubule length is important for allowing rapid remodeling of the cytoskeleton and efficient T cell polarization. The immune system is composed of many types of cells that can recognize foreign molecules and pathogens so they can eliminate them. When cells in the body become infected with a pathogen, they can process the pathogen’s proteins and present them on their own surface. Specialized immune cells can then recognize infected cells and interact with them, forming an ‘immunological synapse’. These synapses play an important role in immune response: they activate the immune system and allow it to kill harmful cells. To form an immunological synapse, an immune cell must reorganize its internal contents, including an aster-shaped scaffold made of tiny protein tubes called microtubules. The center of this scaffold moves towards the immunological synapse as it forms. This re-orientation of the microtubules towards the immunological synapse is known as 'polarization' and it happens very rapidly, but it is not yet clear how it works. One molecule involved in the polarization process is called KIF21B, a protein that can walk along microtubules, building up at the ends and affecting their growth. Whether KIF21B makes microtubules grow more quickly, or more slowly, is a matter of debate, and the impact microtubule length has on immunological synapse formation is unknown. Here, Hooikaas, Damstra et al. deleted the gene for KIF21B from human immune cells called T cells to find out how it affected their ability to form an immunological synapse. Without KIF21B, the T cells grew microtubules that were longer than normal, and had trouble forming immunological synapses. When the T cells were treated with a drug that stops microtubule growth, their ability to form immunological synapses was restored, suggesting a role for KIF21B. To explore this further, Hooikaas, Damstra et al. replaced the missing KIF21B gene with a gene that coded for a version of the protein that could be seen using microscopy. This revealed that, when KIF21B reaches the ends of microtubules, it stops their growth and triggers their disassembly. Computational modelling showed that cells find it hard to reorient their microtubule scaffolding when the individual tubes are too long. It only takes a small number of KIF21B molecules to shorten the microtubules enough to allow the center of the scaffold to move. Research has linked the KIF21B gene to autoimmune conditions like multiple sclerosis. Microtubules also play an important role in cell division, a critical process driving all types of cancer. Drugs that affect microtubule growth are already available, and a deeper understanding of KIF21B and microtubule regulation in immune cells could help to improve treatments in the future.
Collapse
Affiliation(s)
- Peter Jan Hooikaas
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Hugo Gj Damstra
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Oane J Gros
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Wilhelmina E van Riel
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Maud Martin
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Yesper Th Smits
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Jorg van Loosdregt
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Florian Berger
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
27
|
Bianconi E, Casadei R, Frabetti F, Ventura C, Facchin F, Canaider S. Sex-Specific Transcriptome Differences in Human Adipose Mesenchymal Stem Cells. Genes (Basel) 2020; 11:909. [PMID: 32784482 PMCID: PMC7464371 DOI: 10.3390/genes11080909] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/24/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022] Open
Abstract
In humans, sexual dimorphism can manifest in many ways and it is widely studied in several knowledge fields. It is increasing the evidence that also cells differ according to sex, a correlation still little studied and poorly considered when cells are used in scientific research. Specifically, our interest is on the sex-related dimorphism on the human mesenchymal stem cells (hMSCs) transcriptome. A systematic meta-analysis of hMSC microarrays was performed by using the Transcriptome Mapper (TRAM) software. This bioinformatic tool was used to integrate and normalize datasets from multiple sources and allowed us to highlight chromosomal segments and genes differently expressed in hMSCs derived from adipose tissue (hADSCs) of male and female donors. Chromosomal segments and differentially expressed genes in male and female hADSCs resulted to be related to several processes as inflammation, adipogenic and neurogenic differentiation and cell communication. Obtained results lead us to hypothesize that the donor sex of hADSCs is a variable influencing a wide range of stem cell biologic processes. We believe that it should be considered in biologic research and stem cell therapy.
Collapse
Affiliation(s)
- Eva Bianconi
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)—Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy; (E.B.); (C.V.); (S.C.)
| | - Raffaella Casadei
- Department for Life Quality Studies (QuVi), University of Bologna, Corso D’Augusto 237, 47921 Rimini, Italy;
| | - Flavia Frabetti
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Carlo Ventura
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)—Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy; (E.B.); (C.V.); (S.C.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Federica Facchin
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)—Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy; (E.B.); (C.V.); (S.C.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| | - Silvia Canaider
- National Laboratory of Molecular Biology and Stem Cell Bioengineering of the National Institute of Biostructures and Biosystems (NIBB)—Eldor Lab, at the Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy; (E.B.); (C.V.); (S.C.)
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy;
| |
Collapse
|
28
|
Luthria G, Li R, Wang S, Prytyskach M, Kohler RH, Lauffenburger DA, Mitchison TJ, Weissleder R, Miller MA. In vivo microscopy reveals macrophage polarization locally promotes coherent microtubule dynamics in migrating cancer cells. Nat Commun 2020; 11:3521. [PMID: 32665556 PMCID: PMC7360550 DOI: 10.1038/s41467-020-17147-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/02/2020] [Indexed: 01/07/2023] Open
Abstract
Microtubules (MTs) mediate mitosis, directional signaling, and are therapeutic targets in cancer. Yet in vivo analysis of cancer cell MT behavior within the tumor microenvironment remains challenging. Here we developed an imaging pipeline using plus-end tip tracking and intravital microscopy to quantify MT dynamics in live xenograft tumor models. Among analyzed features, cancer cells in vivo displayed higher coherent orientation of MT dynamics along their cell major axes compared with 2D in vitro cultures, and distinct from 3D collagen gel cultures. This in vivo MT phenotype was reproduced in vitro when cells were co-cultured with IL4-polarized MΦ. MΦ depletion, MT disruption, targeted kinase inhibition, and altered MΦ polarization via IL10R blockade all reduced MT coherence and/or tumor cell elongation. We show that MT coherence is a defining feature for in vivo tumor cell dynamics and migration, modulated by local signaling from pro-tumor macrophages. The regulation of microtubule (MT) dynamics in cancer cells within the tumor microenvironment is less understood. Here, the authors develop an imaging platform to examine MT dynamics in live xenograft models and show that pro-tumor macrophages modulate MT coherence and alignment to promote cancer cell migration.
Collapse
Affiliation(s)
- Gaurav Luthria
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA.,Department of Biomedical Informatics, Harvard Medical School, Boston, MA, 02115, USA
| | - Ran Li
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA.,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Stephanie Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02181, USA
| | - Mark Prytyskach
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
| | - Rainer H Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02181, USA
| | - Timothy J Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA. .,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115, USA. .,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Miles A Miller
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA. .,Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
29
|
Whitelaw JA, Swaminathan K, Kage F, Machesky LM. The WAVE Regulatory Complex Is Required to Balance Protrusion and Adhesion in Migration. Cells 2020; 9:E1635. [PMID: 32646006 PMCID: PMC7407199 DOI: 10.3390/cells9071635] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/15/2022] Open
Abstract
Cells migrating over 2D substrates are required to polymerise actin at the leading edge to form lamellipodia protrusions and nascent adhesions to anchor the protrusion to the substrate. The major actin nucleator in lamellipodia formation is the Arp2/3 complex, which is activated by the WAVE regulatory complex (WRC). Using inducible Nckap1 floxed mouse embryonic fibroblasts (MEFs), we confirm that the WRC is required for lamellipodia formation, and importantly, for generating the retrograde flow of actin from the leading cell edge. The loss of NCKAP1 also affects cell spreading and focal adhesion dynamics. In the absence of lamellipodium, cells can become elongated and move with a single thin pseudopod, which appears devoid of N-WASP. This phenotype was more prevalent on collagen than fibronectin, where we observed an increase in migratory speed. Thus, 2D cell migration on collagen is less dependent on branched actin.
Collapse
Affiliation(s)
| | - Karthic Swaminathan
- CRUK Beatson Institute, Glasgow G61 1BD, UK; (K.S.); (L.M.M.)
- School of Chemistry and Bioscience, University of Bradford, Bradford BD7 1PD, UK
| | - Frieda Kage
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755-3844, USA;
- Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Laura M. Machesky
- CRUK Beatson Institute, Glasgow G61 1BD, UK; (K.S.); (L.M.M.)
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|
30
|
Abstract
Migrating cells need to coordinate extension and retraction of their protrusions to avoid fragmenting. Kopf et al. (2020. J. Cell Biol.https://doi.org/10.1083/jcb.201907154) demonstrate that microtubules help to maintain cell coherence during amoeboid migration by controlling actomyosin contractility in retracting protrusions.
Collapse
Affiliation(s)
- Joyce C.M. Meiring
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Neurobiology, and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
31
|
Abstract
Directed cell migration is critical for embryogenesis and organ development, wound healing and the immune response. Microtubules are dynamic polymers that control directional migration through a number of coordinated processes: microtubules are the tracks for long-distance intracellular transport, crucial for delivery of new membrane components and signalling molecules to the leading edge of a migrating cell and the recycling of adhesion receptors. Microtubules act as force generators and compressive elements to support sustained cell protrusions. The assembly and disassembly of microtubules is coupled to Rho GTPase signalling, thereby controlling actin polymerisation, myosin-driven contractility and the turnover of cellular adhesions locally. Cross-talk of actin and microtubule dynamics is mediated through a number of common binding proteins and regulators. Furthermore, cortical microtubule capture sites are physically linked to focal adhesions, facilitating the delivery of secretory vesicles and efficient cross-talk. Here we summarise the diverse functions of microtubules during cell migration, aiming to show how they contribute to the spatially and temporally coordinated sequence of events that permit efficient, directional and persistent migration.
Collapse
|
32
|
Čermák V, Dostál V, Jelínek M, Libusová L, Kovář J, Rösel D, Brábek J. Microtubule-targeting agents and their impact on cancer treatment. Eur J Cell Biol 2020; 99:151075. [PMID: 32414588 DOI: 10.1016/j.ejcb.2020.151075] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/25/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023] Open
Abstract
Microtubule-targeting agents (MTAs) constitute a diverse group of chemical compounds that bind to microtubules and affect their properties and function. Disruption of microtubules induces various cellular responses often leading to cell cycle arrest or cell death, the most common effect of MTAs. MTAs have found a plethora of practical applications in weed control, as fungicides and antiparasitics, and particularly in cancer treatment. Here we summarize the current knowledge of MTAs, the mechanisms of action and their role in cancer treatment. We further outline the potential use of MTAs in anti-metastatic therapy based on inhibition of cancer cell migration and invasiveness. The two main problems associated with cancer therapy by MTAs are high systemic toxicity and development of resistance. Toxic side effects of MTAs can be, at least partly, eliminated by conjugation of the drugs with various carriers. Moreover, some of the novel MTAs overcome the resistance mediated by both multidrug resistance transporters as well as overexpression of specific β-tubulin types. In anti-metastatic therapy, MTAs should be combined with other drugs to target all modes of cancer cell invasion.
Collapse
Affiliation(s)
- Vladimír Čermák
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Vojtěch Dostál
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic
| | - Michael Jelínek
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lenka Libusová
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic
| | - Jan Kovář
- Department of Biochemistry, Cell and Molecular Biology & Center for Research of Diabetes, Metabolism, and Nutrition, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Daniel Rösel
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic.
| |
Collapse
|
33
|
Abstract
CLIP-associating proteins (CLASPs) form an evolutionarily conserved family of regulatory factors that control microtubule dynamics and the organization of microtubule networks. The importance of CLASP activity has been appreciated for some time, but until recently our understanding of the underlying molecular mechanisms remained basic. Over the past few years, studies of, for example, migrating cells, neuronal development, and microtubule reorganization in plants, along with in vitro reconstitutions, have provided new insights into the cellular roles and molecular basis of CLASP activity. In this Cell Science at a Glance article and the accompanying poster, we will summarize some of these recent advances, emphasizing how they impact our current understanding of CLASP-mediated microtubule regulation.
Collapse
Affiliation(s)
- Elizabeth J Lawrence
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Marija Zanic
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, and Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Luke M Rice
- Department of Biophysics and Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
34
|
Matis M. The Mechanical Role of Microtubules in Tissue Remodeling. Bioessays 2020; 42:e1900244. [PMID: 32249455 DOI: 10.1002/bies.201900244] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/12/2020] [Indexed: 12/31/2022]
Abstract
During morphogenesis, tissues undergo extensive remodeling to get their final shape. Such precise sculpting requires the application of forces generated within cells by the cytoskeleton and transmission of these forces through adhesion molecules within and between neighboring cells. Within individual cells, microtubules together with actomyosin filaments and intermediate filaments form the composite cytoskeleton that controls cell mechanics during tissue rearrangements. While studies have established the importance of actin-based mechanical forces that are coupled via intercellular junctions, relatively little is known about the contribution of other cytoskeletal components such as microtubules to cell mechanics during morphogenesis. In this review the focus is on recent findings, highlighting the direct mechanical role of microtubules beyond its well-established role in trafficking and signaling during tissue formation.
Collapse
Affiliation(s)
- Maja Matis
- Institute of Cell Biology, Medical Faculty, University of Münster, Münster, 48149, Germany.,'Cells in Motion' Interfaculty Centre, University of Münster, Münster, 48149, Germany
| |
Collapse
|
35
|
Firat-Karalar EN. Proximity mapping of the microtubule plus-end tracking protein SLAIN2 using the BioID approach. ACTA ACUST UNITED AC 2020; 44:61-72. [PMID: 32256142 PMCID: PMC7129064 DOI: 10.3906/biy-2002-12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The centrosome is the main microtubule-organizing center of animal cells, which plays key roles in critical cellular processes ranging from cell division to cellular signaling. Accordingly, defects in the structure and function of centrosomes cause various human diseases such as cancer and primary microcephaly. To elucidate the molecular defects underlying these diseases, the biogenesis and functions of the centrosomes have to be fully understood. An essential step towards addressing these questions is the identification and functional dissection of the full repertoire of centrosome proteins. Here, we used high-resolution imaging and showed that the microtubule plus-end tracking protein SLAIN2 localizes to the pericentriolar material at the proximal end of centrioles. To gain insight into its cellular functions and mechanisms, we applied in vivo proximity-dependent biotin identification to SLAIN2 and generated its proximity interaction map. Gene ontology analysis of the SLAIN2 interactome revealed extensive interactions with centriole duplication, ciliogenesis, and microtubule-associated proteins, including previously characterized and uncharacterized interactions. Collectively, our results define SLAIN2 as a component of pericentriolar material and provide an important resource for future studies aimed at elucidating SLAIN2 functions at the centrosome.
Collapse
Affiliation(s)
- Elif Nur Firat-Karalar
- Department of Molecular Biology and Genetics, Faculty of Science, Koç University, İstanbul Turkey
| |
Collapse
|
36
|
Rai A, Liu T, Glauser S, Katrukha EA, Estévez-Gallego J, Rodríguez-García R, Fang WS, Díaz JF, Steinmetz MO, Altmann KH, Kapitein LC, Moores CA, Akhmanova A. Taxanes convert regions of perturbed microtubule growth into rescue sites. NATURE MATERIALS 2020; 19:355-365. [PMID: 31819210 DOI: 10.1038/s41563-019-0546-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 10/24/2019] [Indexed: 06/10/2023]
Abstract
Microtubules are polymers of tubulin dimers, and conformational transitions in the microtubule lattice drive microtubule dynamic instability and affect various aspects of microtubule function. The exact nature of these transitions and their modulation by anticancer drugs such as Taxol and epothilone, which can stabilize microtubules but also perturb their growth, are poorly understood. Here, we directly visualize the action of fluorescent Taxol and epothilone derivatives and show that microtubules can transition to a state that triggers cooperative drug binding to form regions with altered lattice conformation. Such regions emerge at growing microtubule ends that are in a pre-catastrophe state, and inhibit microtubule growth and shortening. Electron microscopy and in vitro dynamics data indicate that taxane accumulation zones represent incomplete tubes that can persist, incorporate tubulin dimers and repeatedly induce microtubule rescues. Thus, taxanes modulate the material properties of microtubules by converting destabilized growing microtubule ends into regions resistant to depolymerization.
Collapse
Affiliation(s)
- Ankit Rai
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Tianyang Liu
- Institute of Structural and Molecular Biology, Birkbeck, University of London, London, UK
| | - Simon Glauser
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | - Eugene A Katrukha
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Juan Estévez-Gallego
- Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Ruddi Rodríguez-García
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Wei-Shuo Fang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Beijing, China
| | - J Fernando Díaz
- Chemical and Physical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, Villigen, Switzerland
- University of Basel, Biozentrum, Basel, Switzerland
| | - Karl-Heinz Altmann
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zürich, Zurich, Switzerland
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Birkbeck, University of London, London, UK
| | - Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
37
|
Brand F, Förster A, Christians A, Bucher M, Thomé CM, Raab MS, Westphal M, Pietsch T, von Deimling A, Reifenberger G, Claus P, Hentschel B, Weller M, Weber RG. FOCAD loss impacts microtubule assembly, G2/M progression and patient survival in astrocytic gliomas. Acta Neuropathol 2020; 139:175-192. [PMID: 31473790 DOI: 10.1007/s00401-019-02067-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 08/12/2019] [Accepted: 08/19/2019] [Indexed: 12/23/2022]
Abstract
In search of novel genes associated with glioma pathogenesis, we have previously shown frequent deletions of the KIAA1797/FOCAD gene in malignant gliomas, and a tumor suppressor function of the encoded focadhesin impacting proliferation and migration of glioma cells in vitro and in vivo. Here, we examined an association of reduced FOCAD gene copy number with overall survival of patients with astrocytic gliomas, and addressed the molecular mechanisms that govern the suppressive effect of focadhesin on glioma growth. FOCAD loss was associated with inferior outcome in patients with isocitrate dehydrogenase 1 or 2 (IDH)-mutant astrocytic gliomas of WHO grades II-IV. Multivariate analysis considering age at diagnosis as well as IDH mutation, MGMT promoter methylation, and CDKN2A/B homozygous deletion status confirmed reduced FOCAD gene copy number as a prognostic factor for overall survival. Using a yeast two-hybrid screen and pull-down assays, tubulin beta-6 and other tubulin family members were identified as novel focadhesin-interacting partners. Tubulins and focadhesin co-localized to centrosomes where focadhesin was enriched in proximity to centrioles. Focadhesin was recruited to microtubules via its interaction partner SLAIN motif family member 2 and reduced microtubule assembly rates, possibly explaining the focadhesin-dependent decrease in cell migration. During the cell cycle, focadhesin levels peaked in G2/M phase and influenced time-dependent G2/M progression potentially via polo like kinase 1 phosphorylation, providing a possible explanation for focadhesin-dependent cell growth reduction. We conclude that FOCAD loss may promote biological aggressiveness and worsen clinical outcome of diffuse astrocytic gliomas by enhancing microtubule assembly and accelerating G2/M phase progression.
Collapse
Affiliation(s)
- Frank Brand
- Department of Human Genetics OE 6300, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Alisa Förster
- Department of Human Genetics OE 6300, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Anne Christians
- Department of Human Genetics OE 6300, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Martin Bucher
- Department of Human Genetics OE 6300, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Carina M Thomé
- Neurology Clinic and National Center for Tumor Diseases, Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marc S Raab
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Manfred Westphal
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Torsten Pietsch
- Department of Neuropathology, University of Bonn Medical School, Bonn, Germany
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Neuropathology, German Consortium for Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Guido Reifenberger
- Department of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Claus
- Department of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Germany
- Center for Systems Neuroscience (ZSN), Hannover, Germany
| | - Bettina Hentschel
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Ruthild G Weber
- Department of Human Genetics OE 6300, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
| |
Collapse
|
38
|
Generation and regulation of microtubule network asymmetry to drive cell polarity. Curr Opin Cell Biol 2019; 62:86-95. [PMID: 31739264 DOI: 10.1016/j.ceb.2019.10.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 01/19/2023]
Abstract
Microtubules control cell architecture by serving as a scaffold for intracellular transport, signaling, and organelle positioning. Microtubules are intrinsically polarized, and their orientation, density, and post-translational modifications both respond and contribute to cell polarity. Animal cells that can rapidly reorient their polarity axis, such as fibroblasts, immune cells, and cancer cells, contain radially organized microtubule arrays anchored at the centrosome and the Golgi apparatus, whereas stably polarized cells often acquire non-centrosomal microtubule networks attached to the cell cortex, nucleus, or other structures. Microtubule density, longevity, and post-translational modifications strongly depend on the dynamics of their plus ends. Factors controlling microtubule plus-end dynamics are often part of cortical assemblies that integrate cytoskeletal organization, cell adhesion, and secretion and are subject to microtubule-dependent feedback regulation. Finally, microtubules can mechanically contribute to cell asymmetry by promoting cell elongation, a property that might be important for cells with dense microtubule arrays growing in soft environments.
Collapse
|
39
|
Parker SS, Krantz J, Kwak EA, Barker NK, Deer CG, Lee NY, Mouneimne G, Langlais PR. Insulin Induces Microtubule Stabilization and Regulates the Microtubule Plus-end Tracking Protein Network in Adipocytes. Mol Cell Proteomics 2019; 18:1363-1381. [PMID: 31018989 PMCID: PMC6601206 DOI: 10.1074/mcp.ra119.001450] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Indexed: 12/21/2022] Open
Abstract
Insulin-stimulated glucose uptake is known to involve microtubules, although the function of microtubules and the microtubule-regulating proteins involved in insulin action are poorly understood. CLASP2, a plus-end tracking microtubule-associated protein (+TIP) that controls microtubule dynamics, was recently implicated as the first +TIP associated with insulin-regulated glucose uptake. Here, using protein-specific targeted quantitative phosphoproteomics within 3T3-L1 adipocytes, we discovered that insulin regulates phosphorylation of the CLASP2 network members G2L1, MARK2, CLIP2, AGAP3, and CKAP5 as well as EB1, revealing the existence of a previously unknown microtubule-associated protein system that responds to insulin. To further investigate, G2L1 interactome studies within 3T3-L1 adipocytes revealed that G2L1 coimmunoprecipitates CLASP2 and CLIP2 as well as the master integrators of +TIP assembly, the end binding (EB) proteins. Live-cell total internal reflection fluorescence microscopy in adipocytes revealed G2L1 and CLASP2 colocalize on microtubule plus-ends. We found that although insulin increases the number of CLASP2-containing plus-ends, insulin treatment simultaneously decreases CLASP2-containing plus-end velocity. In addition, we discovered that insulin stimulates redistribution of CLASP2 and G2L1 from exclusive plus-end tracking to "trailing" behind the growing tip of the microtubule. Insulin treatment increases α-tubulin Lysine 40 acetylation, a mechanism that was observed to be regulated by a counterbalance between GSK3 and mTOR, and led to microtubule stabilization. Our studies introduce insulin-stimulated microtubule stabilization and plus-end trailing of +TIPs as new modes of insulin action and reveal the likelihood that a network of microtubule-associated proteins synergize to coordinate insulin-regulated microtubule dynamics.
Collapse
Affiliation(s)
- Sara S Parker
- From the ‡Department of Cellular & Molecular Medicine
| | - James Krantz
- §Department of Medicine, Division of Endocrinology
| | | | | | - Chris G Deer
- University of Arizona Research Computing, University of Arizona, Tucson, Arizona 85721
| | - Nam Y Lee
- ¶Department of Pharmacology,; ‖Department of Chemistry & Biochemistry, University of Arizona College of Medicine, Tucson, Arizona 85721
| | | | | |
Collapse
|
40
|
van Haren J, Wittmann T. Microtubule Plus End Dynamics - Do We Know How Microtubules Grow?: Cells boost microtubule growth by promoting distinct structural transitions at growing microtubule ends. Bioessays 2019; 41:e1800194. [PMID: 30730055 PMCID: PMC7021488 DOI: 10.1002/bies.201800194] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/22/2018] [Indexed: 12/31/2022]
Abstract
Microtubules form a highly dynamic filament network in all eukaryotic cells. Individual microtubules grow by tubulin dimer subunit addition and frequently switch between phases of growth and shortening. These unique dynamics are powered by GTP hydrolysis and drive microtubule network remodeling, which is central to eukaryotic cell biology and morphogenesis. Yet, our knowledge of the molecular events at growing microtubule ends remains incomplete. Here, recent ultrastructural, biochemical and cell biological data are integrated to develop a realistic model of growing microtubule ends comprised of structurally distinct but biochemically overlapping zones. Proteins that recognize microtubule lattice conformations associated with specific tubulin guanosine nucleotide states may independently control major structural transitions at growing microtubule ends. A model is proposed in which tubulin dimer addition and subsequent closure of the MT wall are optimized in cells to achieve rapid physiological microtubule growth.
Collapse
Affiliation(s)
- Jeffrey van Haren
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
41
|
Zhuang M, Zhao S, Jiang Z, Wang S, Sun P, Quan J, Yan D, Wang X. MALAT1 sponges miR-106b-5p to promote the invasion and metastasis of colorectal cancer via SLAIN2 enhanced microtubules mobility. EBioMedicine 2019; 41:286-298. [PMID: 30797712 PMCID: PMC6444028 DOI: 10.1016/j.ebiom.2018.12.049] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/06/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023] Open
Abstract
Background The low expression of miR93/25 (members of miR-106b~25 cluster) promoted the invasion and metastasis of colon cancer cells, which predicted poor survival. However, the role of miR-106b-5p, the member of miR-106b~25 cluster, in colorectal cancer (CRC) remains unclear. Methods Bioinformatics methods were used to predict the potential pairs of lncRNA-miRNA-mRNA. In situ hybridization and qPCR were used to evaluate the expression of MALAT1 and miR-106b-5p in the paraffin-embedded normal and CRC tissues. Kaplan–Meier analysis with the log-rank test was used for survival analyses. Immunohistochemistry staining was applied to investigate the expression of SLAIN2. Fluorescence recovery after photobleaching assay was applied to observe the microtubule (MT) mobility. In vitro and in vivo invasion and metastasis assays were used to explore the function of MALAT1/miR-106b-5p/SLAIN2 in the progression of CRC. Findings miR-106b-5p was identified as a suppressor in CRC. Functionally, ectopic or silencing the expression of miR-106b-5p inhibited or promoted the invasion and metastasis of CRC cells in vitro and in vivo. The long non-coding RNA MALAT1 regulated the miR-106b-5p expression and further mediated the mobility of SLAIN2-related MTs by functioning as a competing endogenous RNA in vitro and in vivo, which resulted in the progression of CRC. Clinically, low miR-106b-5p expression predicted poor survival of CRC patients, especially in combination with high MALAT1/ SLAIN2 expression. Interpretation miR-106b-5p served as a suppressor in combination with MALAT1/miR-106b-5p/SLAIN2, which might be a group of potential prognostic biomarkers in the prognosis of CRC. Fund This work was supported by National Program Project for Precision Medicine in National Research and Development Plan of China (2016YFC0905300), National Natural Science Foundation of China (81572930), National Key Research and Development Program of the Ministry of Science and Technology of China (2016YFC0905303, 2016YFC1303200), Beijing Science and Technology Program (D17110002617004), Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2018PT32012), CAMS Innovation Fund for Medical Sciences (CIFMS) (2016-I2M-1-001), Incentive Fund for Academic Leaders of Oncology Hospital, Chinese Academy of Medical Sciences (RC2016003), and Beijing Hope Run Special Fund from Cancer Foundation of China (LC2017A19). The project of Shanghai Jiaotong Univversity (YG2017QN30).
Collapse
Affiliation(s)
- Meng Zhuang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Senlin Zhao
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Song Wang
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Peng Sun
- Department of Colorectal Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jichuan Quan
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China
| | - Dongwang Yan
- Department of General Surgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Xishan Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
42
|
Ju RJ, Stehbens SJ, Haass NK. The Role of Melanoma Cell-Stroma Interaction in Cell Motility, Invasion, and Metastasis. Front Med (Lausanne) 2018; 5:307. [PMID: 30460237 PMCID: PMC6232165 DOI: 10.3389/fmed.2018.00307] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/16/2018] [Indexed: 12/21/2022] Open
Abstract
The importance of studying cancer cell invasion is highlighted by the fact that 90% of all cancer-related mortalities are due to metastatic disease. Melanoma metastasis is driven fundamentally by aberrant cell motility within three-dimensional or confined environments. Within this realm of cell motility, cytokines, growth factors, and their receptors are crucial for engaging signaling pathways, which both mediate crosstalk between cancer, stromal, and immune cells in addition to interactions with the surrounding microenvironment. Recently, the study of the mechanical biology of tumor cells, stromal cells and the mechanics of the microenvironment have emerged as important themes in driving invasion and metastasis. While current anti-melanoma therapies target either the MAPK signaling pathway or immune checkpoints, there are no drugs available that specifically inhibit motility and thus invasion and dissemination of melanoma cells during metastasis. One of the reasons for the lack of so-called "migrastatics" is that, despite decades of research, the precise biology of metastatic disease is still not fully understood. Metastatic disease has been traditionally lumped into a single classification, however what is now emergent is that the biology of melanoma metastasis is highly diverse, heterogeneous and exceedingly dynamic-suggesting that not all cases are created equal. The following mini-review discusses melanoma heterogeneity in the context of the emergent theme of mechanobiology and how it influences the tumor-stroma crosstalk during metastasis. Thus, highlighting future therapeutic options for migrastatics and mechanomedicines in the prevention and treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Robert J. Ju
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Samantha J. Stehbens
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Nikolas K. Haass
- The University of Queensland, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
- Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
43
|
|
44
|
Polarized microtubule dynamics directs cell mechanics and coordinates forces during epithelial morphogenesis. Nat Cell Biol 2018; 20:1126-1133. [PMID: 30202051 DOI: 10.1038/s41556-018-0193-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 08/09/2018] [Indexed: 11/08/2022]
Abstract
Coordinated rearrangements of cytoskeletal structures are the principal source of forces that govern cell and tissue morphogenesis1,2. However, unlike for actin-based mechanical forces, our knowledge about the contribution of forces originating from other cytoskeletal components remains scarce. Here, we establish microtubules as central components of cell mechanics during tissue morphogenesis. We find that individual cells are mechanically autonomous during early Drosophila wing epithelium development. Each cell contains a polarized apical non-centrosomal microtubule cytoskeleton that bears compressive forces, whereby acute elimination of microtubule-based forces leads to cell shortening. We further establish that the Fat planar cell polarity (Ft-PCP) signalling pathway3,4 couples microtubules at adherens junctions (AJs) and patterns microtubule-based forces across a tissue via polarized transcellular stability, thus revealing a molecular mechanism bridging single cell and tissue mechanics. Together, these results provide a physical basis to explain how global patterning of microtubules controls cell mechanics to coordinate collective cell behaviour during tissue remodelling. These results also offer alternative paradigms towards the interplay of contractile and protrusive cytoskeletal forces at the single cell and tissue levels.
Collapse
|
45
|
Actin-Based Cell Protrusion in a 3D Matrix. Trends Cell Biol 2018; 28:823-834. [PMID: 29970282 PMCID: PMC6158345 DOI: 10.1016/j.tcb.2018.06.003] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/01/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Cell migration controls developmental processes (gastrulation and tissue patterning), tissue homeostasis (wound repair and inflammatory responses), and the pathobiology of diseases (cancer metastasis and inflammation). Understanding how cells move in physiologically relevant environments is of major importance, and the molecular machinery behind cell movement has been well studied on 2D substrates, beginning over half a century ago. Studies over the past decade have begun to reveal the mechanisms that control cell motility within 3D microenvironments – some similar to, and some highly divergent from those found in 2D. In this review we focus on migration and invasion of cells powered by actin, including formation of actin-rich protrusions at the leading edge, and the mechanisms that control nuclear movement in cells moving in a 3D matrix. Cell migration has been well studied in 2D, but how this relates to movement in physiological 3D tissues and matrix is not clear, particularly in vertebrate interstitial matrix. In 3D matrix cells actin polymerisation directly contributes to the formation of lamellipodia to facilitate migration and invasion (mesenchymal movement), analogous to 2D migration; actomyosin contractility promotes bleb formation to indirectly promote protrusion (amoeboid movement). Mesenchymal migration can be characterised by polymerisation of actin to form filopodial protrusions, in the absence of lamellipodia. Translocation of the nucleus is emerging as a critical step due to the constrictive environment of 3D matrices, and the mechanisms that transmit force to the nucleus and allow movement are beginning to be uncovered.
Collapse
|
46
|
Aher A, Kok M, Sharma A, Rai A, Olieric N, Rodriguez-Garcia R, Katrukha EA, Weinert T, Olieric V, Kapitein LC, Steinmetz MO, Dogterom M, Akhmanova A. CLASP Suppresses Microtubule Catastrophes through a Single TOG Domain. Dev Cell 2018; 46:40-58.e8. [PMID: 29937387 PMCID: PMC6035287 DOI: 10.1016/j.devcel.2018.05.032] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 04/13/2018] [Accepted: 05/25/2018] [Indexed: 01/05/2023]
Abstract
The dynamic instability of microtubules plays a key role in controlling their organization and function, but the cellular mechanisms regulating this process are poorly understood. Here, we show that cytoplasmic linker-associated proteins (CLASPs) suppress transitions from microtubule growth to shortening, termed catastrophes, including those induced by microtubule-destabilizing agents and physical barriers. Mammalian CLASPs encompass three TOG-like domains, TOG1, TOG2, and TOG3, none of which bind to free tubulin. TOG2 is essential for catastrophe suppression, whereas TOG3 mildly enhances rescues but cannot suppress catastrophes. These functions are inhibited by the C-terminal domain of CLASP2, while the TOG1 domain can release this auto-inhibition. TOG2 fused to a positively charged microtubule-binding peptide autonomously accumulates at growing but not shrinking ends, suppresses catastrophes, and stimulates rescues. CLASPs suppress catastrophes by stabilizing growing microtubule ends, including incomplete ones, preventing their depolymerization and promoting their recovery into complete tubes. TOG2 domain is the key determinant of these activities. CLASPs potently suppress microtubule catastrophes induced by different mechanisms CLASPs act by stabilizing growing microtubule ends, including incomplete ones CLASP2 TOG-like domain, TOG2, is necessary and sufficient for catastrophe inhibition TOG2 fused to a positively charged peptide accumulates at growing microtubule ends
Collapse
Affiliation(s)
- Amol Aher
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Maurits Kok
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands
| | - Ashwani Sharma
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland
| | - Ankit Rai
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Natacha Olieric
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland
| | - Ruddi Rodriguez-Garcia
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Eugene A Katrukha
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Tobias Weinert
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland
| | - Vincent Olieric
- Swiss Light Source, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland
| | - Lukas C Kapitein
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institut, CH-5232 Villigen PSI, Switzerland; University of Basel, Biozentrum, 4056 Basel, Switzerland
| | - Marileen Dogterom
- Department of Bionanoscience, Kavli Institute of Nanoscience, Delft University of Technology, van der Maasweg 9, 2629 HZ Delft, the Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands.
| |
Collapse
|
47
|
Seo JH, Agarwal E, Bryant KG, Caino MC, Kim ET, Kossenkov AV, Tang HY, Languino LR, Gabrilovich DI, Cohen AR, Speicher DW, Altieri DC. Syntaphilin Ubiquitination Regulates Mitochondrial Dynamics and Tumor Cell Movements. Cancer Res 2018; 78:4215-4228. [PMID: 29898993 DOI: 10.1158/0008-5472.can-18-0595] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/30/2018] [Accepted: 06/06/2018] [Indexed: 02/06/2023]
Abstract
Syntaphilin (SNPH) inhibits the movement of mitochondria in tumor cells, preventing their accumulation at the cortical cytoskeleton and limiting the bioenergetics of cell motility and invasion. Although this may suppress metastasis, the regulation of the SNPH pathway is not well understood. Using a global proteomics screen, we show that SNPH associates with multiple regulators of ubiquitin-dependent responses and is ubiquitinated by the E3 ligase CHIP (or STUB1) on Lys111 and Lys153 in the microtubule-binding domain. SNPH ubiquitination did not result in protein degradation, but instead anchored SNPH on tubulin to inhibit mitochondrial motility and cycles of organelle fusion and fission, that is dynamics. Expression of ubiquitination-defective SNPH mutant Lys111→Arg or Lys153→Arg increased the speed and distance traveled by mitochondria, repositioned mitochondria to the cortical cytoskeleton, and supported heightened tumor chemotaxis, invasion, and metastasis in vivo Interference with SNPH ubiquitination activated mitochondrial dynamics, resulting in increased recruitment of the fission regulator dynamin-related protein-1 (Drp1) to mitochondria and Drp1-dependent tumor cell motility. These data uncover nondegradative ubiquitination of SNPH as a key regulator of mitochondrial trafficking and tumor cell motility and invasion. In this way, SNPH may function as a unique, ubiquitination-regulated suppressor of metastasis.Significance: These findings reveal a new mechanism of metastasis suppression by establishing the role of SNPH ubiquitination in inhibiting mitochondrial dynamics, chemotaxis, and metastasis. Cancer Res; 78(15); 4215-28. ©2018 AACR.
Collapse
Affiliation(s)
- Jae Ho Seo
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Ekta Agarwal
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Kelly G Bryant
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - M Cecilia Caino
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Eui Tae Kim
- Division of Cancer Pathobiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Andrew V Kossenkov
- Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania
| | - Hsin-Yao Tang
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania.,Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania.,Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Dmitry I Gabrilovich
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania.,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Andrew R Cohen
- Department of Electrical and Computer Engineering, College of Engineering, Drexel University, Philadelphia, Pennsylvania
| | - David W Speicher
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania.,Center for Systems and Computational Biology, The Wistar Institute, Philadelphia, Pennsylvania.,Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, Pennsylvania
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, The Wistar Institute, Philadelphia, Pennsylvania. .,Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania
| |
Collapse
|
48
|
Abstract
How do the cells in our body reconfigure their shape to achieve complex tasks like migration and mitosis, yet maintain their shape in response to forces exerted by, for instance, blood flow and muscle action? Cell shape control is defined by a delicate mechanical balance between active force generation and passive material properties of the plasma membrane and the cytoskeleton. The cytoskeleton forms a space-spanning fibrous network comprising three subsystems: actin, microtubules and intermediate filaments. Bottom-up reconstitution of minimal synthetic cells where these cytoskeletal subsystems are encapsulated inside a lipid vesicle provides a powerful avenue to dissect the force balance that governs cell shape control. Although encapsulation is technically demanding, a steady stream of advances in this technique has made the reconstitution of shape-changing minimal cells increasingly feasible. In this topical review we provide a route-map of the recent advances in cytoskeletal encapsulation techniques and outline recent reports that demonstrate shape change phenomena in simple biomimetic vesicle systems. We end with an outlook toward the next steps required to achieve more complex shape changes with the ultimate aim of building a fully functional synthetic cell with the capability to autonomously grow, divide and move.
Collapse
Affiliation(s)
- Yuval Mulla
- These authors contributed equally to this work
| | | | | |
Collapse
|
49
|
Martin M, Veloso A, Wu J, Katrukha EA, Akhmanova A. Control of endothelial cell polarity and sprouting angiogenesis by non-centrosomal microtubules. eLife 2018; 7:33864. [PMID: 29547120 PMCID: PMC5898915 DOI: 10.7554/elife.33864] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 03/13/2018] [Indexed: 12/11/2022] Open
Abstract
Microtubules control different aspects of cell polarization. In cells with a radial microtubule system, a pivotal role in setting up asymmetry is attributed to the relative positioning of the centrosome and the nucleus. Here, we show that centrosome loss had no effect on the ability of endothelial cells to polarize and move in 2D and 3D environments. In contrast, non-centrosomal microtubules stabilized by the microtubule minus-end-binding protein CAMSAP2 were required for directional migration on 2D substrates and for the establishment of polarized cell morphology in soft 3D matrices. CAMSAP2 was also important for persistent endothelial cell sprouting during in vivo zebrafish vessel development. In the absence of CAMSAP2, cell polarization in 3D could be partly rescued by centrosome depletion, indicating that in these conditions the centrosome inhibited cell polarity. We propose that CAMSAP2-protected non-centrosomal microtubules are needed for establishing cell asymmetry by enabling microtubule enrichment in a single-cell protrusion. Networks of blood vessels grow like trees. Sprouts appear on existing vessels, stretching out to form new branches in a process called angiogenesis. The cells responsible are the same cells that line the finished vessels. These “endothelial cells” start the process by reorganizing themselves to face the direction of the new sprout, changing shape to become asymmetrical, and then they begin to migrate. Beneath the surface, a network of protein scaffolding supports each migrating cell. The scaffolding includes tube-like fibers called microtubules that extend towards the cell membrane and organize the inside of the cell. Destroying microtubules damages blood vessel formation, but their exact role remains unclear. A structure called the centrosome can organize microtubules within cells. The centrosome was generally believed to act like a compass, pointing in the direction that the cell will move. Microtubules can anchor to the centrosome, and this structure is thought to play an important role in cell migration. Yet, many microtubules organize without it; these microtubules instead are organized by a compartment of the cell called the Golgi apparatus and are stabilized by a protein named CAMSAP2. Martin et al. now report that removing the cells’ centrosomes did not affect cell migration, but getting rid of CAMSAP2 did. Analysis of cell shape and movement in cells grown in the laboratory and in living animals revealed that cells cannot become asymmetrical, or “polarize”, and migrate without CAMSAP2. In a two-dimensional wound-healing assay, a sheet of cells originally grown from the vessels of a human umbilical cord was scratched, and a microscope was then used to record the cell’s movement as they repaired the injury. Normally, the cells on either side move in a straight line using their microtubules, and though the process was not affected in cells without centrosomes, it was in those without CAMSAP2. Even more striking results were seen in three-dimensional assays. When the same blood vessel cells from human umbilical cords are grown as spheres inside collagen gels, they form sprouts as they would in the body. Without CAMSAP2, the cells could not organize their microtubules and they were unable to elongate in one direction and form stable sprouts. Lastly, depleting CAMSAP2 also prevented the normal formation of blood vessels in zebrafish embryos. Taken together, these findings change our understanding of how microtubules affect cell movement and how important the centrosome is for this process. Further work could have an impact on human health, not least in cancer research. Tumors need a good blood supply to grow, so understanding how to block blood vessel formation could lead to new treatments. Microtubules are already a target for cancer therapy, so future work could help to optimize the use of existing drugs.
Collapse
Affiliation(s)
- Maud Martin
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Alexandra Veloso
- Interdisciplinary Cluster for Applied Genoproteomics, University of Liège, Liège, Belgium.,GIGA-Molecular Biology in Diseases, University of Liège, Liège, Belgium
| | - Jingchao Wu
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Eugene A Katrukha
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
50
|
Development and validation of a gene expression-based signature to predict distant metastasis in locoregionally advanced nasopharyngeal carcinoma: a retrospective, multicentre, cohort study. Lancet Oncol 2018; 19:382-393. [DOI: 10.1016/s1470-2045(18)30080-9] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/14/2017] [Accepted: 11/29/2017] [Indexed: 02/02/2023]
|