1
|
Lei X, Sun E, Ru X, Quan Y, Chen X, Zhang Q, Lu Y, Huang Q, Chen Y, Li W, Feng H, Yang Y, Hu R. Acetylation of α-tubulin restores endothelial cell injury and blood-brain barrier disruption after intracerebral hemorrhage in mice. Exp Mol Med 2025:10.1038/s12276-025-01454-9. [PMID: 40335634 DOI: 10.1038/s12276-025-01454-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 05/09/2025] Open
Abstract
Damage to endothelial cells (ECs) is a key factor in blood-brain barrier (BBB) disruption after intracerebral hemorrhage (ICH). While microtubules are essential for EC structure, their role in BBB injury remains unclear. Here we investigated the role of acetylated α-tubulin (α-Ac-Tub) in BBB integration after ICH. Using an autologous blood injection model in the striatum, we showed that the expression of α-Ac-Tub and MEC17, an α-tubulin acetyltransferase, significantly decreased along the vessels around the hematoma after ICH. Conditional MEC17 knockout in ECs further reduced α-Ac-Tub levels and exacerbated BBB leakage, brain edema, hematoma expansion, inflammation and motor dysfunction. Conversely, selective α-Ac-Tub upregulation in ECs via intravenous delivery of AAV-BI30-MEC17-GFP alleviated BBB dysfunction and improved motor recovery. Similarly, the HDAC6 inhibitor tubastatin A enhanced α-Ac-Tub levels, mitigating BBB damage and neurological deficits. Mechanistically, α-Ac-Tub deficiency in ECs reduced tight junction proteins (ZO-1 and Claudin5) and increased F-actin stress fibers through RhoA activation. Together, our findings highlighted α-Ac-Tub as a therapeutic target for restoring BBB function and reducing brain injury after ICH.
Collapse
Affiliation(s)
- Xuejiao Lei
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Eryi Sun
- Department of Neurosurgery, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Xufang Ru
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yulian Quan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuezhu Chen
- Department of Pathology, Public Health Medical Center, Chongqing, China
| | - Qian Zhang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yougling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wenyan Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China.
| | - Yang Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China.
- Department of Neurosurgery, The 904th Hospital of PLA, Anhui Medical University, Wuxi, China.
| | - Rong Hu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
2
|
Abdellatef SA, Wang H, Nakanishi J. Microtubules Disruption Alters the Cellular Structures and Mechanics Depending on Underlying Chemical Cues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2312282. [PMID: 39344221 PMCID: PMC11962689 DOI: 10.1002/smll.202312282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 09/19/2024] [Indexed: 10/01/2024]
Abstract
The extracellular matrix determines cell morphology and stiffness by manipulating the cytoskeleton. The impacts of extracellular matrix cues, including the mechanical and topographical cues on microtubules and their role in biological behaviors, are previously studied. However, there is a lack of understanding about how microtubules (MTs) are affected by environmental chemical cues, such as extracellular matrix density. Specifically, it is crucial to understand the connection between cellular morphology and mechanics induced by chemical cues and the role of microtubules in these cellular responses. To address this, surfaces with high and low cRGD (cyclic Arginine-Glycine-Aspartic acid) peptide ligand densities are used. The cRGD is diluted with a bioinert ligand to prevent surface native cellular remodeling. The cellular morphology, actin, and microtubules differ on these surfaces. Confocal fluorescence microscopes and atomic force microscopy (AFM) are used to determine the structural and mechanical cellular responses with and without microtubules. Microtubules are vital as an intracellular scaffold in elongated morphology correlated with low cRGD compared to rounded morphology in high cRGD substrates. The contributions of MTs to nucleus morphology and cellular mechanics are based on the underlying cRGD densities. Finally, this study reveals a significant correlation between MTs, actin networks, and vimentin in response to the underlying densities of cRGD.
Collapse
Affiliation(s)
- Shimaa A. Abdellatef
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
| | - Hongxin Wang
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
| | - Jun Nakanishi
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
- Graduate School of Advanced EngineeringTokyo University of Science6‐3‐1, NiijukuKatsushika‐kuTokyo125‐8585Japan
- Graduate School of Advanced Science and EngineeringWaseda University3‐4‐1 OkuboShinjuku‐kuTokyo169‐8555Japan
| |
Collapse
|
3
|
Uesugi K, Obata S, Nagayama K. Micro tensile tester measurement of biomechanical properties and adhesion force of microtubule-polymerization-inhibited cancer cells. J Mech Behav Biomed Mater 2024; 156:106586. [PMID: 38805872 DOI: 10.1016/j.jmbbm.2024.106586] [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: 03/08/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 05/30/2024]
Abstract
Both mechanical and adhesion properties of cancer cells are complex and reciprocally related to migration, invasion, and metastasis with large cell deformation. Therefore, we evaluated these properties for human cervical cancer cells (HeLa) simultaneously using our previously developed micro tensile tester system. For efficient evaluation, we developed image analysis software to modify the system. The software can analyze the tensile force in real time. The modified system can evaluate the tensile stiffness of cells to which a large deformation is applied, also evaluate the adhesion strength of cancer cells that adhered to a culture substrate and were cultured for several days with their adhesion maturation. We used the modified system to simultaneously evaluate the stiffness of the cancer cells to which a large deformation was applied and their adhesion strength. The obtained results revealed that the middle phase of tensile stiffness and adhesion force of the microtubule-depolymerized group treated with colchicine (an anti-cancer drug) (stiffness, 13.4 ± 7.5 nN/%; adhesion force, 460.6 ± 258.2 nN) were over two times larger than those of the control group (stiffness, 5.0 ± 3.5 nN/%; adhesion force, 168.2 ± 98.0 nN). Additionally, the same trend was confirmed with the detailed evaluation of cell surface stiffness using an atomic force microscope. Confocal fluorescence microscope observations showed that the stress fibers (SFs) of colchicine-treated cells were aligned in the same direction, and focal adhesions (FAs) of the cells developed around both ends of the SFs and aligned parallel to the developed direction of the SFs. There was a possibility that the microtubule depolymerization by the colchicine treatment induced the development of SFs and FAs and subsequently caused an increment of cell stiffness and adhesion force. From the above results, we concluded the modified system would be applicable to cancer detection and anti-cancer drug efficacy tests.
Collapse
Affiliation(s)
- Kaoru Uesugi
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan
| | - Shota Obata
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan
| | - Kazuaki Nagayama
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan.
| |
Collapse
|
4
|
Mao Y, Wickström SA. Mechanical state transitions in the regulation of tissue form and function. Nat Rev Mol Cell Biol 2024; 25:654-670. [PMID: 38600372 DOI: 10.1038/s41580-024-00719-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 04/12/2024]
Abstract
From embryonic development, postnatal growth and adult homeostasis to reparative and disease states, cells and tissues undergo constant changes in genome activity, cell fate, proliferation, movement, metabolism and growth. Importantly, these biological state transitions are coupled to changes in the mechanical and material properties of cells and tissues, termed mechanical state transitions. These mechanical states share features with physical states of matter, liquids and solids. Tissues can switch between mechanical states by changing behavioural dynamics or connectivity between cells. Conversely, these changes in tissue mechanical properties are known to control cell and tissue function, most importantly the ability of cells to move or tissues to deform. Thus, tissue mechanical state transitions are implicated in transmitting information across biological length and time scales, especially during processes of early development, wound healing and diseases such as cancer. This Review will focus on the biological basis of tissue-scale mechanical state transitions, how they emerge from molecular and cellular interactions, and their roles in organismal development, homeostasis, regeneration and disease.
Collapse
Affiliation(s)
- Yanlan Mao
- Laboratory for Molecular Cell Biology, University College London, London, UK.
- Institute for the Physics of Living Systems, University College London, London, UK.
| | - Sara A Wickström
- Department of Cell and Tissue Dynamics, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Helsinki Institute of Life Science, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
5
|
Tobin MP, Pfeifer CR, Zhu PK, Hayes BH, Wang M, Vashisth M, Xia Y, Phan SH, Belt SA, Irianto J, Discher DE. Differences in cell shape, motility, and growth reflect chromosomal number variations that can be visualized with live-cell ChReporters. Mol Biol Cell 2023; 34:br19. [PMID: 37903225 PMCID: PMC10848937 DOI: 10.1091/mbc.e23-06-0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 09/15/2023] [Accepted: 10/10/2023] [Indexed: 11/01/2023] Open
Abstract
Chromosome numbers often change dynamically in tumors and cultured cells, which complicates therapy as well as understanding genotype-mechanotype relationships. Here we use a live-cell "ChReporter" method to identify cells with a single chromosomal loss in efforts to better understand differences in cell shape, motility, and growth. We focus on a standard cancer line and first show clonal populations that retain the ChReporter exhibit large differences in cell and nuclear morphology as well as motility. Phenotype metrics follow simple rules, including migratory persistence scaling with speed, and cytoskeletal differences are evident from drug responses, imaging, and single-cell RNA sequencing. However, mechanotype-genotype relationships between fluorescent ChReporter-positive clones proved complex and motivated comparisons of clones that differ only in loss or retention of a Chromosome-5 ChReporter. When lost, fluorescence-null cells show low expression of Chromosome-5 genes, including a key tumor suppressor APC that regulates microtubules and proliferation. Colonies are compact, nuclei are rounded, and cells proliferate more, with drug results implicating APC, and patient survival data indicating an association in multiple tumor-types. Visual identification of genotype with ChReporters can thus help clarify mechanotype and mechano-evolution.
Collapse
Affiliation(s)
- Michael P. Tobin
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | | | | | - Brandon H. Hayes
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Mai Wang
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Manasvita Vashisth
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Yuntao Xia
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Steven H. Phan
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Susanna A. Belt
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Jerome Irianto
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Dennis E. Discher
- Mol. Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
6
|
Zhang X, Shu S, Feng Z, Qiu Y, Bao H, Zhu Z. Microtubule stabilization promotes the synthesis of type 2 collagen in nucleus pulposus cell by activating hippo-yap pathway. Front Pharmacol 2023; 14:1102318. [PMID: 36778003 PMCID: PMC9909034 DOI: 10.3389/fphar.2023.1102318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/18/2023] [Indexed: 01/27/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is the cardinal pathological mechanism that underlies low back pain. Mechanical stress of the intervertebral disc may result in a change in nucleus pulposus cells state, matrix degradation, and degeneration of the disc. Microtubules, which are components of the cytoskeleton, are involved in driving or regulating signal pathways, which sense and transmit mechano-transduction. Microtubule and the related proteins play an important role in the development of many diseases, while little is known about the role of microtubules in nucleus pulposus cells. Researchers have found that type II collagen (COL2) expression is promoted by microtubule stabilization in synovial mesenchymal stem cells. In this study, we demonstrated that microtubule stabilization promotes the expression of COL2 in nucleus pulposus cells. Stabilized microtubules stimulating Hippo signaling pathway, inhibiting YAP protein expression and activity. In addition, microtubules stabilization promotes the expression of COL2 and alleviates disc degeneration in rats. In summary, our study for the first time, identifies microtubule as a promising therapeutic target for IDD, up-regulating the synthesis of COL2 via Hippo-Yap pathway. Our findings may provide new insights into the etiologies and pathology for IDD, further, targeting of microtubule acetylation may be an effective strategy for the treatment of IDD.
Collapse
Affiliation(s)
| | | | | | | | - Hongda Bao
- *Correspondence: Hongda Bao, ; Zezhang Zhu,
| | | |
Collapse
|
7
|
Bär SI, Dittmer A, Nitzsche B, Ter-Avetisyan G, Fähling M, Klefenz A, Kaps L, Biersack B, Schobert R, Höpfner M. Chimeric HDAC and the cytoskeleton inhibitor broxbam as a novel therapeutic strategy for liver cancer. Int J Oncol 2022; 60:73. [PMID: 35485292 PMCID: PMC9097774 DOI: 10.3892/ijo.2022.5363] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 02/11/2022] [Indexed: 12/24/2022] Open
Abstract
Broxbam, also known as N-hydroxy-4-{1-methoxy-4-[4′-(3′-bromo-4′,5′-dimethoxyphenyl)-oxazol-5′-yl]-2-phenoxy} butanamide, is a novel chimeric inhibitor that contains two distinct pharmacophores in its molecular structure. It has been previously demonstrated to inhibit the activity of histone deacetylases (HDAC) and tubulin polymerisation, two critical components required for cancer growth and survival. In the present study, the potential suitability of broxbam for the treatment of liver cancer was investigated. The effects of broxbam on cell proliferation and apoptosis, in addition to the under-lying molecular mechanism of action, were first investigated in primary liver cancer cell lines Huh7, HepG2, TFK1 and EGI1. Real-time proliferation measurements made using the iCEL-Ligence system and viable cell number counting following crystal violet staining) revealed that broxbam time- and dose-dependently reduced the proliferation of liver cancer cell lines with IC50 values <1 µM. In addition, a significant inhibition of the growth of hepatoblastoma microtumours on the chorioallantoic membranes (CAM) of fertilised chicken eggs by broxbam was observed according to results from the CAM assay, suggesting antineoplastic potency in vivo. Broxbam also exerted apoptotic effects through p53- and mitochondria-driven caspase-3 activation in Huh7 and HepG2 cells according to data from western blotting (p53 and phosphorylated p53), mitochondrial membrane potential measurements (JC-1 assay) and fluorometric capsase-3 measurements. Notably, no contribution of unspecific cytotoxic effects mediated by broxbam were observed from LDH-release measurements. HDAC1, -2, -4 and -6 expression was measured by western blotting and the HDAC inhibitory potency of broxbam was next evaluated using subtype-specific HDAC enzymatic assays, which revealed a largely pan-HDAC inhibitory activity with the most potent inhibition observed on HDAC6. Silencing HDAC6 expression in Huh7 cells led to a drop in the expression of the proliferation markers Ki-67 and E2F3, suggesting that HDAC6 inhibition by broxbam may serve a predomi-nant role in their antiproliferative effects on liver cancer cells. Immunofluorescence staining of cytoskeletal proteins (α-tubulin & actin) of broxbam-treated HepG2 cells revealed a pronounced inhibition of tubulin polymerisation, which was accompanied by reduced cell migration as determined by wound healing scratch assays. Finally, data from zebrafish angiogenesis assays revealed marked antiangiogenic effects of broxbam in vivo, as shown by the suppression of subintestinal vein growth in zebrafish embryos. To conclude, the pleiotropic anticancer activities of this novel chimeric HDAC- and tubulin inhibitor broxbam suggest that this compound is a promising candidate for liver cancer treatment, which warrants further pre-clinical and clinical evaluation.
Collapse
Affiliation(s)
- Sofia Isolde Bär
- Organic Chemistry Laboratory, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Alexandra Dittmer
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| | - Bianca Nitzsche
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| | - Gohar Ter-Avetisyan
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| | - Michael Fähling
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| | - Adrian Klefenz
- Institute of Translational Immunology, University Medical Center of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Leonard Kaps
- Institute of Translational Immunology, University Medical Center of the Johannes Gutenberg University, D-55131 Mainz, Germany
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Rainer Schobert
- Organic Chemistry Laboratory, University of Bayreuth, D-95447 Bayreuth, Germany
| | - Michael Höpfner
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, D-10117 Berlin, Germany
| |
Collapse
|
8
|
Seetharaman S, Vianay B, Roca V, Farrugia AJ, De Pascalis C, Boëda B, Dingli F, Loew D, Vassilopoulos S, Bershadsky A, Théry M, Etienne-Manneville S. Microtubules tune mechanosensitive cell responses. NATURE MATERIALS 2022; 21:366-377. [PMID: 34663953 DOI: 10.1038/s41563-021-01108-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 08/20/2021] [Indexed: 05/05/2023]
Abstract
Mechanotransduction is a process by which cells sense the mechanical properties of their surrounding environment and adapt accordingly to perform cellular functions such as adhesion, migration and differentiation. Integrin-mediated focal adhesions are major sites of mechanotransduction and their connection with the actomyosin network is crucial for mechanosensing as well as for the generation and transmission of forces onto the substrate. Despite having emerged as major regulators of cell adhesion and migration, the contribution of microtubules to mechanotransduction still remains elusive. Here, we show that talin- and actomyosin-dependent mechanosensing of substrate rigidity controls microtubule acetylation (a tubulin post-translational modification) by promoting the recruitment of α-tubulin acetyltransferase 1 (αTAT1) to focal adhesions. Microtubule acetylation tunes the mechanosensitivity of focal adhesions and Yes-associated protein (YAP) translocation. Microtubule acetylation, in turn, promotes the release of the guanine nucleotide exchange factor GEF-H1 from microtubules to activate RhoA, actomyosin contractility and traction forces. Our results reveal a fundamental crosstalk between microtubules and actin in mechanotransduction that contributes to mechanosensitive cell adhesion and migration.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Université Paris Descartes, Paris, France
| | - Benoit Vianay
- Paris University, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, Paris, France
| | - Vanessa Roca
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Aaron J Farrugia
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Chiara De Pascalis
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Batiste Boëda
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Florent Dingli
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | - Damarys Loew
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | | | - Alexander Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Manuel Théry
- Paris University, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
9
|
Seetharaman S, Vianay B, Roca V, Farrugia AJ, De Pascalis C, Boëda B, Dingli F, Loew D, Vassilopoulos S, Bershadsky A, Théry M, Etienne-Manneville S. Microtubules tune mechanosensitive cell responses. NATURE MATERIALS 2022; 21:366-377. [PMID: 34663953 DOI: 10.1101/2020.07.22.205203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 08/20/2021] [Indexed: 05/24/2023]
Abstract
Mechanotransduction is a process by which cells sense the mechanical properties of their surrounding environment and adapt accordingly to perform cellular functions such as adhesion, migration and differentiation. Integrin-mediated focal adhesions are major sites of mechanotransduction and their connection with the actomyosin network is crucial for mechanosensing as well as for the generation and transmission of forces onto the substrate. Despite having emerged as major regulators of cell adhesion and migration, the contribution of microtubules to mechanotransduction still remains elusive. Here, we show that talin- and actomyosin-dependent mechanosensing of substrate rigidity controls microtubule acetylation (a tubulin post-translational modification) by promoting the recruitment of α-tubulin acetyltransferase 1 (αTAT1) to focal adhesions. Microtubule acetylation tunes the mechanosensitivity of focal adhesions and Yes-associated protein (YAP) translocation. Microtubule acetylation, in turn, promotes the release of the guanine nucleotide exchange factor GEF-H1 from microtubules to activate RhoA, actomyosin contractility and traction forces. Our results reveal a fundamental crosstalk between microtubules and actin in mechanotransduction that contributes to mechanosensitive cell adhesion and migration.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Université Paris Descartes, Paris, France
| | - Benoit Vianay
- Paris University, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, Paris, France
| | - Vanessa Roca
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Aaron J Farrugia
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Chiara De Pascalis
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Batiste Boëda
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Florent Dingli
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | - Damarys Loew
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | | | - Alexander Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Manuel Théry
- Paris University, INSERM, CEA, Hôpital Saint Louis, Institut Universitaire d'Hematologie, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
10
|
Cyclic stretching-induced epithelial cell reorientation is driven by microtubule-modulated transverse extension during the relaxation phase. Sci Rep 2021; 11:14803. [PMID: 34285275 PMCID: PMC8292395 DOI: 10.1038/s41598-021-93987-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/29/2021] [Indexed: 11/29/2022] Open
Abstract
Many types of adherent cells are known to reorient upon uniaxial cyclic stretching perpendicularly to the direction of stretching to facilitate such important events as wound healing, angiogenesis, and morphogenesis. While this phenomenon has been documented for decades, the underlying mechanism remains poorly understood. Using an on-stage stretching device that allowed programmable stretching with synchronized imaging, we found that the reorientation of NRK epithelial cells took place primarily during the relaxation phase when cells underwent rapid global retraction followed by extension transverse to the direction of stretching. Inhibition of myosin II caused cells to orient along the direction of stretching, whereas disassembly of microtubules enhanced transverse reorientation. Our results indicate distinct roles of stretching and relaxation in cell reorientation and implicate a role of myosin II-dependent contraction via a microtubule-modulated mechanism. The importance of relaxation phase also explains the difference between the responses to cyclic and static stretching.
Collapse
|
11
|
Kubiak A, Chighizola M, Schulte C, Bryniarska N, Wesołowska J, Pudełek M, Lasota M, Ryszawy D, Basta-Kaim A, Laidler P, Podestà A, Lekka M. Stiffening of DU145 prostate cancer cells driven by actin filaments - microtubule crosstalk conferring resistance to microtubule-targeting drugs. NANOSCALE 2021; 13:6212-6226. [PMID: 33885607 DOI: 10.1039/d0nr06464e] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The crucial role of microtubules in the mitotic-related segregation of chromosomes makes them an excellent target for anticancer microtubule targeting drugs (MTDs) such as vinflunine (VFL), colchicine (COL), and docetaxel (DTX). MTDs affect mitosis by directly perturbing the structural organisation of microtubules. By a direct assessment of the biomechanical properties of prostate cancer DU145 cells exposed to different MTDs using atomic force microscopy, we show that cell stiffening is a response to the application of all the studied MTDs (VFL, COL, DTX). Changes in cellular rigidity are typically attributed to remodelling of the actin filaments in the cytoskeleton. Here, we demonstrate that cell stiffening can be driven by crosstalk between actin filaments and microtubules in MTD-treated cells. Our findings improve the interpretation of biomechanical data obtained for living cells in studies of various physiological and pathological processes.
Collapse
Affiliation(s)
- Andrzej Kubiak
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Kraków, Poland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Gryadunova AA, Koudan EV, Rodionov SA, Pereira FDAS, Meteleva NY, Kasyanov VA, Parfenov VA, Kovalev AV, Khesuani YD, Mironov VA, Bulanova EA. Cytoskeleton systems contribute differently to the functional intrinsic properties of chondrospheres. Acta Biomater 2020; 118:141-152. [PMID: 33045401 DOI: 10.1016/j.actbio.2020.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
Cytoskeleton systems, actin microfilaments, microtubules (MTs) and intermediate filaments (IFs) provide the biomechanical stability and spatial organization in cells. To understand the specific contributions of each cytoskeleton systems to intrinsic properties of spheroids, we've scrutinized the effects of the cytoskeleton perturbants, cytochalasin D (Cyto D), nocodazole (Noc) and withaferin A (WFA) on fusion, spreading on adhesive surface, morphology and biomechanics of chondrospheres (CSs). We confirmed that treatment with Cyto D but not with Noc or WFA severely affected CSs fusion and spreading dynamics and significantly reduced biomechanical properties of cell aggregates. Noc treatment affected spheroids spreading but not the fusion and surprisingly enhanced their stiffness. Vimentin intermediate filaments (VIFs) reorganization affected CSs spreading only. The analysis of all three cytoskeleton systems contribution to spheroids intrinsic properties was performed for the first time.
Collapse
Affiliation(s)
- Anna A Gryadunova
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation; Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russian Federation.
| | - Elizaveta V Koudan
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation.
| | - Sergey A Rodionov
- N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, Moscow 127299, Russian Federation
| | - F D A S Pereira
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation
| | - Nina Yu Meteleva
- I.D. Papanin Institute for Biology of Inland Waters RAS, Borok 152742, Russian Federation
| | - Vladimir A Kasyanov
- Riga Stradins University, Riga LV-1007, Latvia; Riga Technical University, Riga LV-1658, Latvia
| | - Vladislav A Parfenov
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation
| | - Alexey V Kovalev
- N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, Moscow 127299, Russian Federation
| | - Yusef D Khesuani
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation
| | - Vladimir A Mironov
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation; Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russian Federation
| | - Elena A Bulanova
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation.
| |
Collapse
|
13
|
Fine N, Gracey E, Dimitriou I, La Rose J, Glogauer M, Rottapel R. GEF-H1 Is Required for Colchicine Inhibition of Neutrophil Rolling and Recruitment in Mouse Models of Gout. THE JOURNAL OF IMMUNOLOGY 2020; 205:3300-3310. [PMID: 33199537 DOI: 10.4049/jimmunol.1900783] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/20/2020] [Indexed: 11/19/2022]
Abstract
Gout is a painful arthritic inflammatory disease caused by buildup of monosodium urate (MSU) crystals in the joints. Colchicine, a microtubule-depolymerizing agent that is used in prophylaxis and treatment of acute gout flare, alleviates the painful inflammatory response to MSU crystals. Using i.p. and intra-articular mouse models of gout-like inflammation, we found that GEF-H1/GEF-H1/AHRGEF2, a microtubule-associated Rho-GEF, was necessary for the inhibitory effect of colchicine on neutrophil recruitment. GEF-H1 was required for neutrophil polarization in response to colchicine, characterized by uropod formation, accumulation of F-actin and myosin L chain at the leading edge, and accumulation of phosphorylated myosin L chain, flotillin-2, and P-selectin glycoprotein ligand-1 (PSGL-1) in the uropod. Wild-type neutrophils that were pre-exposed to colchicine failed to roll or accumulate on activated endothelial monolayers, whereas GEF-H1 knockout (GEF-H1-/-) neutrophils were unaffected by treatment with colchicine. In vivo, colchicine blocked MSU-induced recruitment of neutrophils to the peritoneum and the synovium in wild-type mice, but not in GEF-H1-/- mice. Inhibition of macrophage IL-1β production by colchicine was independent of GEF-H1, supporting a neutrophil-intrinsic mode of action. Our results suggest that the anti-inflammatory effects of colchicine in acute gout-like inflammation can be accounted for by inhibition of neutrophil-rolling interactions with the inflamed vasculature and occurs through GEF-H1-dependent neutrophil stimulation by colchicine. These results contribute to our understanding of the therapeutic action of colchicine, and could inform the application of this drug in other conditions.
Collapse
Affiliation(s)
- Noah Fine
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| | - Eric Gracey
- Vlaams Institute for Biotechnology Centre for Inflammation Research, 9052 Ghent, Belgium.,Department of Internal Medicine and Pediatrics, University of Ghent, 9000 Ghent, Belgium
| | - Ioannis Dimitriou
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - José La Rose
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, Ontario M5G 1G6, Canada
| | - Robert Rottapel
- Department of Immunology, Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario M5G 1L7, Canada; .,Department of Medicine, Ontario Institute for Cancer Research, University of Toronto, Toronto, Ontario M5G 1L7, Canada; and.,Division of Rheumatology, St. Michael's Hospital, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
14
|
Traverso S. Anxiety and depression: A matter of stiffness? Med Hypotheses 2020; 145:110344. [PMID: 33075584 DOI: 10.1016/j.mehy.2020.110344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/06/2020] [Accepted: 10/07/2020] [Indexed: 10/23/2022]
Abstract
Cells react to stress by the universal responses of "fluidization" or "reinforcement" (stiffening) of the cytoplasm, through dramatic re-arrangements of the cytoskeleton. Here it is suggested that, at a supracellular level, the brain exhibits such a fundamental behavior as part of its complex response to stress: it is hypothesized that the soft gel formed by brain cell cytoskeletons and the surrounding extracellular matrix (the "cytoskeletons-matrix system") undergoes transitions either to sol (fluidization) or stiff gel (reinforcement) as a very fundamental and evolutionarily conserved brain response to stress, alongside more sophisticated neural pathways. Sol state corresponds to increased cell activity (a sort of "fight or flight" response), whereas stiff gel state corresponds to inactivity (an "immobility" strategy). Psychological stress, through simple stress signals such as pH changes, would lead to an initial tissue fluidization in key regions of the brain, followed, if the stress stimuli persist, by reinforcement (slow formation of actomyosin stress fibers and matrix stiffening). It is also hypothesized that the cytoskeletons-matrix system is one of the biological correlates of so-called "background feelings", i.e conscious feelings built on inner chemical-physical states of the body. Optimal dynamics of the cytoskeletons-matrix system would contribute to a core feeling of well-being, while shifts towards fluidization (activation) or stiffening (inactivation) would contribute to background feelings at the basis of anxiety and stress-induced depression, respectively. It is suggested that the cytoskeletons-matrix system behaves as a "self-organized critical system", anxiety and depression arising whenever the system is driven too far from the optimal critical point. Finally, some application hints from the proposed ideas are given.
Collapse
|
15
|
Tumor Treating Fields (TTFields) Hinder Cancer Cell Motility through Regulation of Microtubule and Acting Dynamics. Cancers (Basel) 2020; 12:cancers12103016. [PMID: 33080774 PMCID: PMC7603026 DOI: 10.3390/cancers12103016] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Tumor Treating Fields (TTFields), encompassing alternating electric fields within the intermediate frequency range, is an anticancer treatment delivered to the tumor region through transducer arrays placed non-invasively on the skin. Although established as an anti-mitotic treatment modality, the anti-metastatic potential of TTFields and their effect on rapid cytoskeletal dynamics during cellular motility warrant further investigation. In this study, we report that TTFields application induces changes in microtubule organization leading to interference with the directionality and robustness of cancer cell migration. We show that these changes in microtubule organization result in activation of GEF-H1/RhoA/ROCK signaling pathway, and the consequent formation of focal adhesions and changes in actin cytoskeleton architecture. Together, these results propose a novel mechanism by which TTFields induce changes in microtubule and actin organization and dynamics, thereby disrupting processes important for polarity generation and motility in cancer cells. Abstract Tumor Treating Fields (TTFields) are noninvasive, alternating electric fields within the intermediate frequency range (100–300 kHz) that are utilized as an antimitotic cancer treatment. TTFields are loco-regionally delivered to the tumor region through 2 pairs of transducer arrays placed on the skin. This novel treatment modality has been FDA-approved for use in patients with glioblastoma and malignant pleural mesothelioma based on clinical trial data demonstrating efficacy and safety; and is currently under investigation in other types of solid tumors. TTFields were shown to induce an anti-mitotic effect by exerting bi-directional forces on highly polar intracellular elements, such as tubulin and septin molecules, eliciting abnormal microtubule polymerization during spindle formation as well as aberrant cleavage furrow formation. Previous studies have demonstrated that TTFields inhibit metastatic properties in cancer cells. However, the consequences of TTFields application on cytoskeleton dynamics remain undetermined. In this study, methods utilized in combination to study the effects of TTFields on cancer cell motility through regulation of microtubule and actin dynamics included confocal microscopy, computational tools, and biochemical analyses. Mechanisms by which TTFields treatment disrupted cellular polarity were (1) interference with microtubule assembly and directionality; (2) altered regulation of Guanine nucleotide exchange factor-H1 (GEF-H1), Ras homolog family member A (RhoA), and Rho-associated coiled-coil kinase (ROCK) activity; and (3) induced formation of radial protrusions of peripheral actin filaments and focal adhesions. Overall, these data identified discrete effects of TTFields that disrupt processes crucial for cancer cell motility.
Collapse
|
16
|
Regulation of invadosomes by microtubules: Not only a matter of railways. Eur J Cell Biol 2020; 99:151109. [DOI: 10.1016/j.ejcb.2020.151109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022] Open
|
17
|
Swiatlowska P, Sanchez-Alonso JL, Mansfield C, Scaini D, Korchev Y, Novak P, Gorelik J. Short-term angiotensin II treatment regulates cardiac nanomechanics via microtubule modifications. NANOSCALE 2020; 12:16315-16329. [PMID: 32720664 DOI: 10.1039/d0nr02474k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Mechanical properties of single myocytes contribute to the whole heart performance, but the measurement of mechanics in living cells at high resolution with minimal force interaction remains challenging. Angiotensin II (AngII) is a peptide hormone that regulates a number of physiological functions, including heart performance. It has also been shown to contribute to cell mechanics by inducing cell stiffening. Using non-contact high-resolution Scanning Ion Conductance Microscopy (SICM), we determine simultaneously cell topography and membrane transverse Young's modulus (YM) by a constant pressure application through a nanopipette. While applying pressure, the vertical position is recorded and a deformation map is generated from which YM can be calculated and corrected for the uneven geometry. High resolution of this method also allows studying specific membrane subdomains, such as Z-grooves and crests. We found that short-term AngII treatment reduces the transversal YM in isolated adult rat cardiomyocytes acting via an AT1 receptor. Blocking either a TGF-β1 receptor or Rho kinase abolishes this effect. Analysis of the cytoskeleton showed that AngII depletes microtubules by decreasing long-lived detyrosinated and acetylated microtubule populations. Interestingly, in the failing cardiomyocytes, which are stiffer than controls, the short-term AngII treatment also reduces the YM, thus normalizing the mechanical state of cells. This suggests that the short-term softening effect of AngII on cardiac cells is opposite to the well-characterized long-term hypertrophic effect. In conclusion, we generate a precise nanoscale indication map of location-specific transverse cortical YM within the cell and this can substantially advance our understanding of cellular mechanics in a physiological environment, for example in isolated cardiac myocytes.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| | - Jose L Sanchez-Alonso
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| | - Catherine Mansfield
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| | - Denis Scaini
- Department of Medicine, Imperial College London, London, UK and International School for Advanced Studies, Trieste, Italy
| | - Yuri Korchev
- Department of Medicine, Imperial College London, London, UK and Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Pavel Novak
- Department of Medicine, Imperial College London, London, UK and National University of Science and Technology, MISiS, Leninskiy prospect 4, Moscow, 119991, Russia
| | - Julia Gorelik
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
18
|
Zheng Y, Gong J, Zhen Y. Focal adhesion kinase is activated by microtubule-depolymerizing agents and regulates membrane blebbing in human endothelial cells. J Cell Mol Med 2020; 24:7228-7238. [PMID: 32452639 PMCID: PMC7339229 DOI: 10.1111/jcmm.15273] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/01/2020] [Accepted: 03/27/2020] [Indexed: 12/17/2022] Open
Abstract
Microtubule-depolymerizing agents can selectively disrupt tumor vessels via inducing endothelial membrane blebbing. However, the mechanism regulating blebbing is largely unknown. IMB5046 is a newly discovered microtubule-depolymerizing agent. Here, the functions of focal adhesion kinase (FAK) during IMB5046-induced blebbing and the relevant mechanism are studied. We found that IMB5046 induced membrane blebbing and reassembly of focal adhesions in human vascular endothelial cells. Both FAK inhibitor and knock-down expression of FAK inhibited IMB5046-induced blebbing. Mechanism study revealed that IMB5046 induced the activation of FAK via GEF-H1/ Rho/ ROCK/ MLC2 pathway. cRGD peptide, a ligand of integrin, also blocked IMB5046-induced blebbing. After activation, FAK further promoted the phosphorylation of MLC2. This positive feedback loop caused more intensive actomyosin contraction and continuous membrane blebbing. FAK inhibitor blocked membrane blebbing via inhibiting actomyosin contraction, and stimulated stress fibre formation via promoting the phosphorylation of HSP27. Conclusively, these results demonstrate that FAK is a molecular switch controlling endothelial blebbing and stress fibre formation. Our study provides a new molecular mechanism for microtubule-depolymerizing agents to be used as vascular disrupting agents.
Collapse
Affiliation(s)
- Yan‐Bo Zheng
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jian‐Hua Gong
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yong‐Su Zhen
- Institute of Medicinal BiotechnologyChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
19
|
Galigniana NM, Charó NL, Uranga R, Cabanillas AM, Piwien-Pilipuk G. Oxidative stress induces transcription of telomeric repeat-containing RNA (TERRA) by engaging PKA signaling and cytoskeleton dynamics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118643. [DOI: 10.1016/j.bbamcr.2020.118643] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 12/27/2019] [Accepted: 01/02/2020] [Indexed: 12/11/2022]
|
20
|
Effect of F-actin and Microtubules on Cellular Mechanical Behavior Studied Using Atomic Force Microscope and an Image Recognition-Based Cytoskeleton Quantification Approach. Int J Mol Sci 2020; 21:ijms21020392. [PMID: 31936268 PMCID: PMC7014474 DOI: 10.3390/ijms21020392] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 11/16/2022] Open
Abstract
Cytoskeleton morphology plays a key role in regulating cell mechanics. Particularly, cellular mechanical properties are directly regulated by the highly cross-linked and dynamic cytoskeletal structure of F-actin and microtubules presented in the cytoplasm. Although great efforts have been devoted to investigating the qualitative relation between the cellular cytoskeleton state and cell mechanical properties, comprehensive quantification results of how the states of F-actin and microtubules affect mechanical behavior are still lacking. In this study, the effect of both F-actin and microtubules morphology on cellular mechanical properties was quantified using atomic force microscope indentation experiments together with the proposed image recognition-based cytoskeleton quantification approach. Young’s modulus and diffusion coefficient of NIH/3T3 cells with different cytoskeleton states were quantified at different length scales. It was found that the living NIH/3T3 cells sense and adapt to the F-actin and microtubules states: both the cellular elasticity and poroelasticity are closely correlated to the depolymerization degree of F-actin and microtubules at all measured indentation depths. Moreover, the significance of the quantitative effects of F-actin and microtubules in affecting cellular mechanical behavior is depth-dependent.
Collapse
|
21
|
Tonazzini I, Van Woerden GM, Masciullo C, Mientjes EJ, Elgersma Y, Cecchini M. The role of ubiquitin ligase E3A in polarized contact guidance and rescue strategies in UBE3A-deficient hippocampal neurons. Mol Autism 2019; 10:41. [PMID: 31798818 PMCID: PMC6884852 DOI: 10.1186/s13229-019-0293-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 10/17/2019] [Indexed: 11/10/2022] Open
Abstract
Background Although neuronal extracellular sensing is emerging as crucial for brain wiring and therefore plasticity, little is known about these processes in neurodevelopmental disorders. Ubiquitin protein ligase E3A (UBE3A) plays a key role in neurodevelopment. Lack of UBE3A leads to Angelman syndrome (AS), while its increase is among the most prevalent genetic causes of autism (e.g., Dup15q syndrome). By using microstructured substrates that can induce specific directional stimuli in cells, we previously found deficient topographical contact guidance in AS neurons, which was linked to a dysregulated activation of the focal adhesion pathway. Methods Here, we study axon and dendrite contact guidance and neuronal morphological features of wild-type, AS, and UBE3A-overexpressing neurons (Dup15q autism model) on micrograting substrates, with the aim to clarify the role of UBE3A in neuronal guidance. Results We found that loss of axonal contact guidance is specific for AS neurons while UBE3A overexpression does not affect neuronal directional polarization along microgratings. Deficits at the level of axonal branching, growth cone orientation and actin fiber content, focal adhesion (FA) effectors, and actin fiber-binding proteins were observed in AS neurons. We tested different rescue strategies for restoring correct topographical guidance in AS neurons on microgratings, by either UBE3A protein re-expression or by pharmacological treatments acting on cytoskeleton contractility. Nocodazole, a drug that depolymerizes microtubules and increases cell contractility, rescued AS axonal alignment to the gratings by partially restoring focal adhesion pathway activation. Surprisingly, UBE3A re-expression only resulted in partial rescue of the phenotype. Conclusions We identified a specific in vitro deficit in axonal topographical guidance due selectively to the loss of UBE3A, and we further demonstrate that this defective guidance can be rescued to a certain extent by pharmacological or genetic treatment strategies. Overall, cytoskeleton dynamics emerge as important partners in UBE3A-mediated contact guidance responses. These results support the view that UBE3A-related deficits in early neuronal morphogenesis may lead to defective neuronal connectivity and plasticity.
Collapse
Affiliation(s)
- Ilaria Tonazzini
- Istituto Nanoscienze- Consiglio Nazionale delle Ricerche (CNR) & Scuola Normale Superiore, NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
- Department of Neuroscience, ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Wytemaweg 80, 3000 CA Rotterdam, the Netherlands
| | - Geeske M. Van Woerden
- Department of Neuroscience, ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Wytemaweg 80, 3000 CA Rotterdam, the Netherlands
| | - Cecilia Masciullo
- Istituto Nanoscienze- Consiglio Nazionale delle Ricerche (CNR) & Scuola Normale Superiore, NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Edwin J. Mientjes
- Department of Neuroscience, ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Wytemaweg 80, 3000 CA Rotterdam, the Netherlands
| | - Ype Elgersma
- Department of Neuroscience, ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC, Wytemaweg 80, 3000 CA Rotterdam, the Netherlands
| | - Marco Cecchini
- Istituto Nanoscienze- Consiglio Nazionale delle Ricerche (CNR) & Scuola Normale Superiore, NEST, Piazza San Silvestro 12, 56127 Pisa, Italy
| |
Collapse
|
22
|
Targeting the IL-1β/EHD1/TUBB3 axis overcomes resistance to EGFR-TKI in NSCLC. Oncogene 2019; 39:1739-1755. [DOI: 10.1038/s41388-019-1099-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/24/2022]
|
23
|
Weber A, Iturri J, Benitez R, Zemljic-Jokhadar S, Toca-Herrera JL. Microtubule disruption changes endothelial cell mechanics and adhesion. Sci Rep 2019; 9:14903. [PMID: 31624281 PMCID: PMC6797797 DOI: 10.1038/s41598-019-51024-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/24/2019] [Indexed: 12/27/2022] Open
Abstract
The interest in studying the mechanical and adhesive properties of cells has increased in recent years. The cytoskeleton is known to play a key role in cell mechanics. However, the role of the microtubules in shaping cell mechanics is not yet well understood. We have employed Atomic Force Microscopy (AFM) together with confocal fluorescence microscopy to determine the role of microtubules in cytomechanics of Human Umbilical Vein Endothelial Cells (HUVECs). Additionally, the time variation of the adhesion between tip and cell surface was studied. The disruption of microtubules by exposing the cells to two colchicine concentrations was monitored as a function of time. Already, after 30 min of incubation the cells stiffened, their relaxation times increased (lower fluidity) and the adhesion between tip and cell decreased. This was accompanied by cytoskeletal rearrangements, a reduction in cell area and changes in cell shape. Over the whole experimental time, different behavior for the two used concentrations was found while for the control the values remained stable. This study underlines the role of microtubules in shaping endothelial cell mechanics.
Collapse
Affiliation(s)
- Andreas Weber
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 11, A-1190, Vienna, Austria.
| | - Jagoba Iturri
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 11, A-1190, Vienna, Austria
| | - Rafael Benitez
- Dpto. Matemáticas para la Economía y la Empresa, Facultad de Economía, Universidad de Valencia, Avda. Tarongers s/n, 46022, Valencia, Spain
| | - Spela Zemljic-Jokhadar
- Department of Biophysics, Medicine Faculty, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - José L Toca-Herrera
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 11, A-1190, Vienna, Austria.
| |
Collapse
|
24
|
Seetharaman S, Etienne-Manneville S. Microtubules at focal adhesions – a double-edged sword. J Cell Sci 2019; 132:132/19/jcs232843. [DOI: 10.1242/jcs.232843] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
ABSTRACT
Cell adhesion to the extracellular matrix is essential for cellular processes, such as migration and invasion. In response to cues from the microenvironment, integrin-mediated adhesions alter cellular behaviour through cytoskeletal rearrangements. The tight association of the actin cytoskeleton with adhesive structures has been extensively studied, whereas the microtubule network in this context has gathered far less attention. In recent years, however, microtubules have emerged as key regulators of cell adhesion and migration through their participation in adhesion turnover and cellular signalling. In this Review, we focus on the interactions between microtubules and integrin-mediated adhesions, in particular, focal adhesions and podosomes. Starting with the association of microtubules with these adhesive structures, we describe the classical role of microtubules in vesicular trafficking, which is involved in the turnover of cell adhesions, before discussing how microtubules can also influence the actin–focal adhesion interplay through RhoGTPase signalling, thereby orchestrating a very crucial crosstalk between the cytoskeletal networks and adhesions.
Collapse
Affiliation(s)
- Shailaja Seetharaman
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France
- Université Paris Descartes, Center for Research and Interdisciplinarity, Sorbonne Paris Cité, 12 Rue de l'École de Médecine, 75006 Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Equipe Labellisée Ligue Contre le Cancer, 75015 Paris, France
| |
Collapse
|
25
|
Sengupta S, Rothenberg KE, Li H, Hoffman BD, Bursac N. Altering integrin engagement regulates membrane localization of K ir2.1 channels. J Cell Sci 2019; 132:jcs225383. [PMID: 31391240 PMCID: PMC6771140 DOI: 10.1242/jcs.225383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 07/31/2019] [Indexed: 12/26/2022] Open
Abstract
How ion channels localize and distribute on the cell membrane remains incompletely understood. We show that interventions that vary cell adhesion proteins and cell size also affect the membrane current density of inward-rectifier K+ channels (Kir2.1; encoded by KCNJ2) and profoundly alter the action potential shape of excitable cells. By using micropatterning to manipulate the localization and size of focal adhesions (FAs) in single HEK293 cells engineered to stably express Kir2.1 channels or in neonatal rat cardiomyocytes, we establish a robust linear correlation between FA coverage and the amplitude of Kir2.1 current at both the local and whole-cell levels. Confocal microscopy showed that Kir2.1 channels accumulate in membrane proximal to FAs. Selective pharmacological inhibition of key mediators of protein trafficking and the spatially dependent alterations in the dynamics of Kir2.1 fluorescent recovery after photobleaching revealed that the Kir2.1 channels are transported to the cell membrane uniformly, but are preferentially internalized by endocytosis at sites that are distal from FAs. Based on these results, we propose adhesion-regulated membrane localization of ion channels as a fundamental mechanism of controlling cellular electrophysiology via mechanochemical signals, independent of the direct ion channel mechanogating.
Collapse
Affiliation(s)
- Swarnali Sengupta
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | | | - Hanjun Li
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| |
Collapse
|
26
|
Müller MT, Schempp R, Lutz A, Felder T, Felder E, Miklavc P. Interaction of microtubules and actin during the post-fusion phase of exocytosis. Sci Rep 2019; 9:11973. [PMID: 31427591 PMCID: PMC6700138 DOI: 10.1038/s41598-019-47741-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 07/09/2019] [Indexed: 01/24/2023] Open
Abstract
Exocytosis is the intracellular trafficking step where a secretory vesicle fuses with the plasma membrane to release vesicle content. Actin and microtubules both play a role in exocytosis; however, their interplay is not understood. Here we study the interaction of actin and microtubules during exocytosis in lung alveolar type II (ATII) cells that secrete surfactant from large secretory vesicles. Surfactant extrusion is facilitated by an actin coat that forms on the vesicle shortly after fusion pore opening. Actin coat compression allows hydrophobic surfactant to be released from the vesicle. We show that microtubules are localized close to actin coats and stay close to the coats during their compression. Inhibition of microtubule polymerization by colchicine and nocodazole affected the kinetics of actin coat formation and the extent of actin polymerisation on fused vesicles. In addition, microtubule and actin cross-linking protein IQGAP1 localized to fused secretory vesicles and IQGAP1 silencing influenced actin polymerisation after vesicle fusion. This study demonstrates that microtubules can influence actin coat formation and actin polymerization on secretory vesicles during exocytosis.
Collapse
Affiliation(s)
- M Tabitha Müller
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Rebekka Schempp
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Anngrit Lutz
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Tatiana Felder
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Edward Felder
- Institute of General Physiology, Ulm University, Albert-Einstein Allee 11, 89081, Ulm, Germany
| | - Pika Miklavc
- School of Environment and Life Sciences, University of Salford, The Crescent, M54WT, Salford, United Kingdom.
| |
Collapse
|
27
|
Rafiq NBM, Nishimura Y, Plotnikov SV, Thiagarajan V, Zhang Z, Shi S, Natarajan M, Viasnoff V, Kanchanawong P, Jones GE, Bershadsky AD. A mechano-signalling network linking microtubules, myosin IIA filaments and integrin-based adhesions. NATURE MATERIALS 2019; 18:638-649. [PMID: 31114072 DOI: 10.1038/s41563-019-0371-y] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 04/10/2019] [Indexed: 06/09/2023]
Abstract
The interrelationship between microtubules and the actin cytoskeleton in mechanoregulation of integrin-mediated adhesions is poorly understood. Here, we show that the effects of microtubules on two major types of cell-matrix adhesion, focal adhesions and podosomes, are mediated by KANK family proteins connecting the adhesion protein talin with microtubule tips. Both total microtubule disruption and microtubule uncoupling from adhesions by manipulations with KANKs trigger a massive assembly of myosin IIA filaments, augmenting focal adhesions and disrupting podosomes. Myosin IIA filaments are indispensable effectors in the microtubule-driven regulation of integrin-mediated adhesions. Myosin IIA filament assembly depends on Rho activation by the RhoGEF GEF-H1, which is trapped by microtubules when they are connected with integrin-mediated adhesions via KANK proteins but released after their disconnection. Thus, microtubule capture by integrin-mediated adhesions modulates the GEF-H1-dependent effect of microtubules on the assembly of myosin IIA filaments. Subsequent actomyosin reorganization then remodels the focal adhesions and podosomes, closing the regulatory loop.
Collapse
Affiliation(s)
- Nisha Bte Mohd Rafiq
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK
| | - Yukako Nishimura
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Sergey V Plotnikov
- Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | | | - Zhen Zhang
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Shidong Shi
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | | | - Virgile Viasnoff
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- CNRS UMI 3639, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Pakorn Kanchanawong
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Deparment of Biomedical Engineering, National University of Singapore, Singapore, Singapore.
| | - Gareth E Jones
- Randall Centre for Cell & Molecular Biophysics, King's College London, London, UK.
| | - Alexander D Bershadsky
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore.
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
28
|
Fine N, Dimitriou ID, Rottapel R. Go with the flow: GEF-H1 mediated shear stress mechanotransduction in neutrophils. Small GTPases 2017; 11:23-31. [PMID: 29188751 DOI: 10.1080/21541248.2017.1332505] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Neutrophils in circulation experience significant shear forces due to blood flow when they tether to the vascular endothelium. Biochemical and biophysical responses of neutrophils to the physical force of flowing blood modulate their behavior and promote tissue recruitment under pro-inflammatory conditions. Neutrophil mechanotransduction responses occur through mechanisms that are not yet fully understood. In our recent work, we showed that GEF-H1, a RhoA specific guanine nucleotide exchange factor (GEF), is required to maintain neutrophil motility and migration in response to shear stress. GEF-H1 re-localizes to flottilin-rich uropods in neutrophils in response to fluid shear stress and promotes spreading and crawling on activated endothelial cells. GEF-H1 drives cellular contractility through myosin light chain (MLC) phosphorylation downstream of the Rho-ROCK signaling axis. We propose that GEF-H1-dependent cell spreading and crawling in shear stress-dependent neutrophil recruitment from the vasculature are due to the specific localization of Rho-induced contractility in the uropod.
Collapse
Affiliation(s)
- Noah Fine
- Matrix Dynamics Group, University of Toronto, Toronto, Ontario, Canada
| | - Ioannis D Dimitriou
- Princess Margaret Cancer Center, Toronto Medical Discovery Tower, Toronto, Ontario, Canada
| | - Robert Rottapel
- Princess Margaret Cancer Center, Toronto Medical Discovery Tower, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
29
|
An engineering insight into the relationship of selective cytoskeletal impairment and biomechanics of HeLa cells. Micron 2017; 102:88-96. [DOI: 10.1016/j.micron.2017.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/22/2017] [Accepted: 09/04/2017] [Indexed: 10/24/2022]
|
30
|
RCCD1 depletion attenuates TGF-β-induced EMT and cell migration by stabilizing cytoskeletal microtubules in NSCLC cells. Cancer Lett 2017; 400:18-29. [DOI: 10.1016/j.canlet.2017.04.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/23/2017] [Accepted: 04/16/2017] [Indexed: 12/22/2022]
|
31
|
Sato Y, Murakami Y, Takahashi M. Semi-retentive cytoskeletal fractionation (SERCYF): A novel method for the biochemical analysis of the organization of microtubule and actin cytoskeleton networks. Biochem Biophys Res Commun 2017; 488:614-620. [PMID: 28526408 DOI: 10.1016/j.bbrc.2017.05.083] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 05/15/2017] [Indexed: 12/27/2022]
Abstract
A variety of biochemical fractionation methods are available for the quantification of cytoskeletal components. However, each method is designed to target only one cytoskeletal network, either the microtubule (MT) or actin cytoskeleton, and non-targeted cytoskeletal networks are ignored. Considering the importance of MT-actin crosstalk, the organization of both the targeted and non-targeted cytoskeletal networks must be retained intact during fractionation for the accurate analysis of cytoskeletal organization. In this study, we reveal that existing fractionation methods, represented by the MT-sedimentation-method for MTs and the Triton X-100 solubility assay-method for actin cytoskeletons, disrupt the organizations of the non-targeted cytoskeletons. We demonstrate a novel fractionation method for the accurate analysis of the cytoskeletal organizations using a taxol-containing PEM-based permeabilization buffer, which we name "semi-retentive cytoskeletal fractionation (SERCYF)-method". The organizations of both MTs and actin cytoskeletons were retained intact even after permeabilization with this buffer. By using the SERCYF-method, we analyzed the effects of nocodazole on the cytoskeletal organizations biochemically and showed promotion of the actin cytoskeletal organization by MT depolymerization.
Collapse
Affiliation(s)
- Yuta Sato
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Yota Murakami
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-8628, Japan; Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| | - Masayuki Takahashi
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo 060-8628, Japan; Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan.
| |
Collapse
|
32
|
Abstract
During an innate immune response, myeloid cells undergo complex morphological adaptations in response to inflammatory cues, which allow them to exit the vasculature, enter the tissues, and destroy invading pathogens. The actin and microtubule cytoskeletons are central to many of the most essential cellular functions including cell division, cell morphology, migration, intracellular trafficking, and signaling. Cytoskeletal structure and regulation are crucial for many myeloid cell functions, which require rapid and dynamic responses to extracellular signals. In this chapter, we review the roles of the actin and microtubule cytoskeletons in myeloid cells, focusing primarily on their roles in chemotaxis and phagocytosis. The role of myeloid cell cytoskeletal defects in hematological disorders is highlighted throughout.
Collapse
|
33
|
Gosselin P, Rando G, Fleury-Olela F, Schibler U. Unbiased identification of signal-activated transcription factors by barcoded synthetic tandem repeat promoter screening (BC-STAR-PROM). Genes Dev 2017; 30:1895-907. [PMID: 27601530 PMCID: PMC5024686 DOI: 10.1101/gad.284828.116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/02/2016] [Indexed: 12/23/2022]
Abstract
Gosselin et al. designed a widely applicable method, dubbed BC-STAR-PROM, to identify signal-activated TFs without any prior knowledge of their properties. To establish proof of concept for BC-STAR-PROM, they applied it to the identification of TFs induced by drugs affecting actin and tubulin cytoskeleton dynamics. The discovery of transcription factors (TFs) controlling pathways in health and disease is of paramount interest. We designed a widely applicable method, dubbed barcorded synthetic tandem repeat promoter screening (BC-STAR-PROM), to identify signal-activated TFs without any a priori knowledge about their properties. The BC-STAR-PROM library consists of ∼3000 luciferase expression vectors, each harboring a promoter (composed of six tandem repeats of synthetic random DNA) and an associated barcode of 20 base pairs (bp) within the 3′ untranslated mRNA region. Together, the promoter sequences encompass >400,000 bp of random DNA, a sequence complexity sufficient to capture most TFs. Cells transfected with the library are exposed to a signal, and the mRNAs that it encodes are counted by next-generation sequencing of the barcodes. This allows the simultaneous activity tracking of each of the ∼3000 synthetic promoters in a single experiment. Here we establish proof of concept for BC-STAR-PROM by applying it to the identification of TFs induced by drugs affecting actin and tubulin cytoskeleton dynamics. BC-STAR-PROM revealed that serum response factor (SRF) is the only immediate early TF induced by both actin polymerization and microtubule depolymerization. Such changes in cytoskeleton dynamics are known to occur during the cell division cycle, and real-time bioluminescence microscopy indeed revealed cell-autonomous SRF–myocardin-related TF (MRTF) activity bouts in proliferating cells.
Collapse
Affiliation(s)
- Pauline Gosselin
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | - Gianpaolo Rando
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| | | | - Ueli Schibler
- Department of Molecular Biology, University of Geneva, CH-1211 Geneva, Switzerland
| |
Collapse
|
34
|
HILI destabilizes microtubules by suppressing phosphorylation and Gigaxonin-mediated degradation of TBCB. Sci Rep 2017; 7:46376. [PMID: 28393858 PMCID: PMC5385498 DOI: 10.1038/srep46376] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 03/15/2017] [Indexed: 02/05/2023] Open
Abstract
Human PIWIL2, aka HILI, is a member of PIWI protein family and overexpresses in various tumors. However, the underlying mechanisms of HILI in tumorigenesis remain largely unknown. TBCB has a critical role in regulating microtubule dynamics and is overexpressed in many cancers. Here we report that HILI inhibits Gigaxonin-mediated TBCB ubiquitination and degradation by interacting with TBCB, promoting the binding between HSP90 and TBCB, and suppressing the interaction between Gigaxonin and TBCB. Meanwhile, HILI can also reduce phosphorylation level of TBCB induced by PAK1. Our results showed that HILI suppresses microtubule polymerization and promotes cell proliferation, migration and invasion via TBCB for the first time, revealing a novel mechanism for HILI in tumorigenesis.
Collapse
|
35
|
Manifacier I, Milan JL, Jeanneau C, Chmilewsky F, Chabrand P, About I. Computational Tension Mapping of Adherent Cells Based on Actin Imaging. PLoS One 2016; 11:e0146863. [PMID: 26812601 PMCID: PMC4728200 DOI: 10.1371/journal.pone.0146863] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 12/21/2015] [Indexed: 01/14/2023] Open
Abstract
Forces transiting through the cytoskeleton are known to play a role in adherent cell activity. Up to now few approaches haves been able to determine theses intracellular forces. We thus developed a computational mechanical model based on a reconstruction of the cytoskeleton of an adherent cell from fluorescence staining of the actin network and focal adhesions (FA). Our custom made algorithm converted the 2D image of an actin network into a map of contractile interactions inside a 2D node grid, each node representing a group of pixels. We assumed that actin filaments observed under fluorescence microscopy, appear brighter when thicker, we thus presumed that nodes corresponding to pixels with higher actin density were linked by stiffer interactions. This enabled us to create a system of heterogeneous interactions which represent the spatial organization of the contractile actin network. The contractility of this interaction system was then adapted to match the level of force the cell truly exerted on focal adhesions; forces on focal adhesions were estimated from their vinculin expressed size. This enabled the model to compute consistent mechanical forces transiting throughout the cell. After computation, we applied a graphical approach on the original actin image, which enabled us to calculate tension forces throughout the cell, or in a particular region or even in single stress fibers. It also enabled us to study different scenarios which may indicate the mechanical role of other cytoskeletal components such as microtubules. For instance, our results stated that the ratio between intra and extra cellular compression is inversely proportional to intracellular tension.
Collapse
Affiliation(s)
- Ian Manifacier
- Aix-Marseille Université, ISM, CNRS, UMR 7287, Marseille, France
- APHM, Institute for Locomotion, Sainte-Marguerite Hospital, 13009, Marseille, France
- * E-mail:
| | - Jean-Louis Milan
- Aix-Marseille Université, ISM, CNRS, UMR 7287, Marseille, France
- APHM, Institute for Locomotion, Sainte-Marguerite Hospital, 13009, Marseille, France
| | | | - Fanny Chmilewsky
- Aix-Marseille Université, ISM, CNRS, UMR 7287, Marseille, France
| | - Patrick Chabrand
- Aix-Marseille Université, ISM, CNRS, UMR 7287, Marseille, France
- APHM, Institute for Locomotion, Sainte-Marguerite Hospital, 13009, Marseille, France
| | - Imad About
- Aix-Marseille Université, ISM, CNRS, UMR 7287, Marseille, France
| |
Collapse
|
36
|
Acharya BR, Espenel C, Libanje F, Raingeaud J, Morgan J, Jaulin F, Kreitzer G. KIF17 regulates RhoA-dependent actin remodeling at epithelial cell-cell adhesions. J Cell Sci 2016; 129:957-70. [PMID: 26759174 DOI: 10.1242/jcs.173674] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 12/31/2015] [Indexed: 12/18/2022] Open
Abstract
The kinesin KIF17 localizes at microtubule plus-ends where it contributes to regulation of microtubule stabilization and epithelial polarization. We now show that KIF17 localizes at cell-cell adhesions and that KIF17 depletion inhibits accumulation of actin at the apical pole of cells grown in 3D organotypic cultures and alters the distribution of actin and E-cadherin in cells cultured in 2D on solid supports. Overexpression of full-length KIF17 constructs or truncation mutants containing the N-terminal motor domain resulted in accumulation of newly incorporated GFP-actin into junctional actin foci, cleared E-cadherin from cytoplasmic vesicles and stabilized cell-cell adhesions to challenge with calcium depletion. Expression of these KIF17 constructs also increased cellular levels of active RhoA, whereas active RhoA was diminished in KIF17-depleted cells. Inhibition of RhoA or its effector ROCK, or expression of LIMK1 kinase-dead or activated cofilin(S3A) inhibited KIF17-induced junctional actin accumulation. Interestingly, KIF17 activity toward actin depends on the motor domain but is independent of microtubule binding. Together, these data show that KIF17 can modify RhoA-GTPase signaling to influence junctional actin and the stability of the apical junctional complex of epithelial cells.
Collapse
Affiliation(s)
- Bipul R Acharya
- Department of Cell and Developmental Biology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Cedric Espenel
- Department of Cell and Developmental Biology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Fotine Libanje
- Gustave Roussy Institute, UMR-8126, 114 rue Edouard Vaillant, Villejuif 94805, France
| | - Joel Raingeaud
- Gustave Roussy Institute, UMR-8126, 114 rue Edouard Vaillant, Villejuif 94805, France
| | - Jessica Morgan
- Department of Cell and Developmental Biology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Fanny Jaulin
- Gustave Roussy Institute, UMR-8126, 114 rue Edouard Vaillant, Villejuif 94805, France
| | - Geri Kreitzer
- Department of Cell and Developmental Biology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
37
|
Nakashima H, Okimura C, Iwadate Y. The molecular dynamics of crawling migration in microtubule-disrupted keratocytes. Biophys Physicobiol 2015; 12:21-9. [PMID: 27493851 PMCID: PMC4736841 DOI: 10.2142/biophysico.12.0_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/24/2015] [Indexed: 12/21/2022] Open
Abstract
Cell-crawling migration plays an essential role in complex biological phenomena. It is now generally believed that many processes essential to such migration are regulated by microtubules in many cells, including fibroblasts and neurons. However, keratocytes treated with nocodazole, which is an inhibitor of microtubule polymerization – and even keratocyte fragments that contain no microtubules – migrate at the same velocity and with the same directionality as normal keratocytes. In this study, we discovered that not only these migration properties, but also the molecular dynamics that regulate such properties, such as the retrograde flow rate of actin filaments, distributions of vinculin and myosin II, and traction forces, are also the same in nocodazole-treated keratocytes as those in untreated keratocytes. These results suggest that microtubules are not in fact required for crawling migration of keratocytes, either in terms of migrating properties or of intracellular molecular dynamics.
Collapse
Affiliation(s)
- Hitomi Nakashima
- Faculty of Science, Yamaguchi University, Yamaguchi 753-8512, Japan
| | - Chika Okimura
- Faculty of Science, Yamaguchi University, Yamaguchi 753-8512, Japan
| | - Yoshiaki Iwadate
- Faculty of Science, Yamaguchi University, Yamaguchi 753-8512, Japan
| |
Collapse
|
38
|
Kunimoto H, Kazama K, Takai M, Oda M, Okada M, Yamawaki H. Chemerin promotes the proliferation and migration of vascular smooth muscle and increases mouse blood pressure. Am J Physiol Heart Circ Physiol 2015; 309:H1017-28. [PMID: 26254337 DOI: 10.1152/ajpheart.00820.2014] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 07/28/2015] [Indexed: 11/22/2022]
Abstract
Blood chemerin concentration shows positive correlation not only with body mass index and serum triglyceride level but also with systolic blood pressure. While it seems likely that chemerin influences vascular smooth muscle cell (SMC) proliferation and migration, which are crucial to the development of hypertension, this remains to be clarified. In the present study, we investigated whether chemerin controls SMC proliferation and migration in vitro and also affects blood pressure in vivo. In vitro, chemerin significantly stimulated rat mesenteric arterial SMC proliferation and migration, as determined by a cell counting assay and Boyden chamber assay, respectively. The migratory effect of chemerin was confirmed in human aortic SMCs. Chemerin significantly increased ROS production in SMCs and phosphorylation of Akt (Ser(473)) and ERK, as measured by fluorescent staining and Western blot analysis, respectively. Various inhibitors (ROS inhibitor: N-acetyl-l-cysteine, phosphatidylinositol 3-kinase inhibitor: LY-294002, MAPKK inhibitor: PD-98059, NADPH oxidase inhibitor: gp91 ds-tat, and xanthine oxidase inhibitor: allopurinol) as well as chemokine-like receptor 1 small interfering RNA significantly inhibited chemerin-induced SMC proliferation and migration. Furthermore, chemerin-neutralizing antibody prevented carotid neointimal hyperplasia in the mouse ligation model. In vivo, chronic chemerin treatment (6 μg/kg, 6 wk) increased systolic blood pressure as well as phosphorylation of Akt and ERK in the mouse isolated aorta. In summary, we, for the first time, demonstrate that chemerin/chemokine-like receptor 1 stimulates SMC proliferation and migration via ROS-dependent phosphorylation of Akt/ERK, which may lead to vascular structural remodeling and an increase in systolic blood pressure.
Collapse
Affiliation(s)
- Hidemizu Kunimoto
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Kyosuke Kazama
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Mizuho Takai
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Mayuko Oda
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| |
Collapse
|
39
|
Li Z, Liu XB, Liu YH, Xue YX, Wang P, Liu LB. Role of cAMP-dependent protein kinase A activity in low-dose endothelial monocyte-activating polypeptide-II-induced opening of blood-tumor barrier. J Mol Neurosci 2014; 56:60-9. [PMID: 25416651 DOI: 10.1007/s12031-014-0467-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 11/11/2014] [Indexed: 01/18/2023]
Abstract
Our previous studies demonstrated that low-dose endothelial monocyte-activating polypeptide-II (EMAP-II) can selectively increase the permeability of blood-tumor barrier (BTB). In addition, low-dose EMAP-II significantly decreases the cyclic adenosine monophosphate (cAMP) concentration and the protein kinase A (PKA) expression level in tumor tissues in the rat C6 glioma model. In this study, an in vitro BTB model was used to investigate the potential role of cAMP/PKA signaling cascade in EMAP-II-induced BTB hyperpermeability. Our data revealed that low-dose EMAP-II (0.05 nM) induced a significant decrease in total intracellular cAMP concentration and PKA activity in rat brain microvascular endothelial cells (RBMECs). Pretreatment with forskolin to increase intracellular cAMP nearly completely blocked the EMAP-II-induced decrease in transendothelial electric resistance and increase in horseradish peroxidase flux across the BTB. Similar pretreatment completely prevented the EMAP-II-induced changes in RhoA/Rho kinase activity, expression and distribution of tight junction-associated protein ZO-1, and myosin light chain phosphorylation, as well as actin cytoskeleton arrangement in RBMECs. Pretreatment with 6Bnz-cAMP to activate PKA significantly attenuated these EMAP-II-induced alterations in RBMECs. In summary, our present study demonstrates that the cAMP/PKA signaling cascade works as a crucial signaling pathway in EMAP-II-induced BTB hyperpermeability.
Collapse
Affiliation(s)
- Zhen Li
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, 110004, Liaoning Province, People's Republic of China,
| | | | | | | | | | | |
Collapse
|
40
|
Goicoechea SM, Awadia S, Garcia-Mata R. I'm coming to GEF you: Regulation of RhoGEFs during cell migration. Cell Adh Migr 2014; 8:535-49. [PMID: 25482524 PMCID: PMC4594598 DOI: 10.4161/cam.28721] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cell migration is a highly regulated multistep process that requires the coordinated regulation of cell adhesion, protrusion, and contraction. These processes require numerous protein–protein interactions and the activation of specific signaling pathways. The Rho family of GTPases plays a key role in virtually every aspect of the cell migration cycle. The activation of Rho GTPases is mediated by a large and diverse family of proteins; the guanine nucleotide exchange factors (RhoGEFs). GEFs work immediately upstream of Rho proteins to provide a direct link between Rho activation and cell–surface receptors for various cytokines, growth factors, adhesion molecules, and G protein-coupled receptors. The regulated targeting and activation of RhoGEFs is essential to coordinate the migratory process. In this review, we summarize the recent advances in our understanding of the role of RhoGEFs in the regulation of cell migration.
Collapse
Key Words
- DH, Dbl-homology
- DHR, DOCK homology region
- DOCK, dedicator of cytokinesis
- ECM, extracellular matrix
- EGF, epidermal growth factor
- FA, focal adhesion
- FN, fibronectin
- GAP, GTPase activating protein
- GDI, guanine nucleotide dissociation inhibitor
- GEF, guanine nucleotide exchange factor
- GPCR, G protein-coupled receptor
- HGF, hepatocyte growth factor
- LPA, lysophosphatidic acid
- MII, myosin II
- PA, phosphatidic acid
- PDGF, platelet-derived growth factor
- PH, pleckstrin-homology
- PIP2, phosphatidylinositol 4, 5-bisphosphate
- PIP3, phosphatidylinositol (3, 4, 5)-trisphosphate.
- Rho GEFs
- Rho GTPases
- bFGF, basic fibroblast growth factor
- cell migration
- cell polarization
- focal adhesions
- guanine nucleotide exchange factors
Collapse
Affiliation(s)
- Silvia M Goicoechea
- a Department of Biological Sciences ; University of Toledo ; Toledo , OH USA
| | | | | |
Collapse
|
41
|
RHGF-1/PDZ-RhoGEF and retrograde DLK-1 signaling drive neuronal remodeling on microtubule disassembly. Proc Natl Acad Sci U S A 2014; 111:16568-73. [PMID: 25359212 DOI: 10.1073/pnas.1410263111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Neurons remodel their connectivity in response to various insults, including microtubule disruption. How neurons sense microtubule disassembly and mount remodeling responses by altering genetic programs in the soma are not well defined. Here we show that in response to microtubule disassembly, the Caenorhabditis elegans PLM neuron remodels by retracting its synaptic branch and overextending the primary neurite. This remodeling required RHGF-1, a PDZ-Rho guanine nucleotide exchange factor (PDZ-RhoGEF) that was associated with and inhibited by microtubules. Independent of the myosin light chain activation, RHGF-1 acted through Rho-dependent kinase LET-502/ROCK and activated a conserved, retrograde DLK-1 MAPK (DLK-1/dual leucine zipper kinase) pathway, which triggered synaptic branch retraction and overgrowth of the PLM neurite in a dose-dependent manner. Our data represent a neuronal remodeling paradigm during development that reshapes the neural circuit by the coordinated removal of the dysfunctional synaptic branch compartment and compensatory extension of the primary neurite.
Collapse
|
42
|
Ramdas NM, Shivashankar GV. Cytoskeletal control of nuclear morphology and chromatin organization. J Mol Biol 2014; 427:695-706. [PMID: 25281900 DOI: 10.1016/j.jmb.2014.09.008] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 09/03/2014] [Accepted: 09/06/2014] [Indexed: 02/06/2023]
Abstract
The nucleus is sculpted toward various morphologies during cellular differentiation and development. Alterations in nuclear shape often result in changes to chromatin organization and genome function. This is thought to be reflective of its role as a cellular mechanotransducer. Recent evidence has highlighted the importance of cytoskeletal organization in defining how nuclear morphology regulates chromatin dynamics. However, the mechanisms underlying cytoskeletal control of chromatin remodeling are not well understood. We demonstrate here the differential influence of perinuclear actin- and microtubule-driven assemblies on nuclear architecture using pharmacological inhibitors and targeted RNA interference knockdown of cytoskeleton components in Drosophila cells. We find evidence that the loss of perinuclear actin assembly results in basolateral enhancement of microtubule organization and this is reflected functionally by enhanced nuclear dynamics. Cytoskeleton reorganization leads to nuclear lamina deformation that influences heterochromatin localization and core histone protein mobility. We also show that modulations in actin-microtubule assembly result in differential gene expression patterns. Taken together, we suggest that perinuclear actin and basolateral microtubule organization exerts mechanical control on nuclear morphology and chromatin dynamics.
Collapse
Affiliation(s)
- Nisha M Ramdas
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560 065, India
| | - G V Shivashankar
- Mechanobiology Institute and Department of Biological Sciences, National University of Singapore, 21 Lower Kent Ridge Road 119077, Singapore.
| |
Collapse
|
43
|
Meng Q, Mongan M, Wang J, Tang X, Zhang J, Kao W, Xia Y. Epithelial sheet movement requires the cooperation of c-Jun and MAP3K1. Dev Biol 2014; 395:29-37. [PMID: 25224220 DOI: 10.1016/j.ydbio.2014.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 08/26/2014] [Accepted: 09/03/2014] [Indexed: 10/24/2022]
Abstract
Epithelial sheet movement is an essential morphogenetic process during mouse embryonic eyelid closure in which Mitogen-Activated Protein 3 Kinase 1 (MAP3K1) and c-Jun play a critical role. Here we show that MAP3K1 associates with the cytoskeleton, activates Jun N-terminal kinase (JNK) and actin polymerization, and promotes the eyelid inferior epithelial cell elongation and epithelium protrusion. Following epithelium protrusion, c-Jun begins to express and acts to promote ERK phosphorylation and migration of the protruding epithelial cells. Homozygous deletion of either gene causes defective eyelid closure, but non-allelic non-complementation does not occur between Map3k1 and c-Jun and the double heterozygotes have normal eyelid closure. Results from this study suggest that MAP3K1 and c-Jun signal through distinct temporal-spatial pathways and that productive epithelium movement for eyelid closure requires the consecutive action of MAP3K1-dependent cytoskeleton reorganization followed by c-Jun-mediated migration.
Collapse
Affiliation(s)
- Qinghang Meng
- Department of Environmental Health, University of Cincinnati, College of Medicine
| | - Maureen Mongan
- Department of Environmental Health, University of Cincinnati, College of Medicine
| | - Jingjing Wang
- Department of Environmental Health, University of Cincinnati, College of Medicine
| | - Xiaofang Tang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center
| | - Jinling Zhang
- Department of Environmental Health, University of Cincinnati, College of Medicine
| | - Winston Kao
- Department of Ophthalmology, University of Cincinnati, College of Medicine
| | - Ying Xia
- Department of Environmental Health, University of Cincinnati, College of Medicine.,Department of Ophthalmology, University of Cincinnati, College of Medicine
| |
Collapse
|
44
|
Huang IH, Hsiao CT, Wu JC, Shen RF, Liu CY, Wang YK, Chen YC, Huang CM, del Álamo JC, Chang ZF, Tang MJ, Khoo KH, Kuo JC. GEF-H1 controls focal adhesion signaling that regulates mesenchymal stem cell lineage commitment. J Cell Sci 2014; 127:4186-200. [PMID: 25107365 PMCID: PMC4179489 DOI: 10.1242/jcs.150227] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Focal adhesions (FAs) undergo maturation that culminates in size and composition changes that modulate adhesion, cytoskeleton remodeling and differentiation. Although it is well recognized that stimuli for osteogenesis of mesenchymal stem cells (MSCs) drive FA maturation, actin organization and stress fiber polarization, the extent to which FA-mediated signals regulated by the FA protein composition specifies MSC commitment remains largely unknown. Here, we demonstrate that, upon dexamethasone (osteogenic induction) treatment, guanine nucleotide exchange factor H1 (GEF-H1, also known as Rho guanine nucleotide exchange factor 2, encoded by ARHGEF2) is significantly enriched in FAs. Perturbation of GEF-H1 inhibits FA formation, anisotropic stress fiber orientation and MSC osteogenesis in an actomyosin-contractility-independent manner. To determine the role of GEF-H1 in MSC osteogenesis, we explore the GEF-H1-modulated FA proteome that reveals non-muscle myosin-II heavy chain-B (NMIIB, also known as myosin-10, encoded by MYH10) as a target of GEF-H1 in FAs. Inhibition of targeting NMIIB into FAs suppresses FA formation, stress fiber polarization, cell stiffness and osteogenic commitments in MSCs. Our data demonstrate a role for FA signaling in specifying MSC commitment.
Collapse
Affiliation(s)
- I-Husan Huang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Cheng-Te Hsiao
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Jui-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Rong-Fong Shen
- Proteomics and Analytical Biochemistry Unit, Research Resources Branch, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Ching-Yi Liu
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan Department of Physiology, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yang-Kao Wang
- Department of Cell Biology and Anatomy, National Cheng Kung University, Tainan 70101, Taiwan Center for Neurotrauma and Neuroregeneration, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Chen Chen
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Chi-Ming Huang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Juan C del Álamo
- Institute of Engineering in Medicine, University of California at San Diego, La Jolla, CA 92093, USA Department of Mechanical and Aerospace Engineering, University of California at San Diego, La Jolla, CA 92093, USA
| | - Zee-Fen Chang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Ming-Jer Tang
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan 70101, Taiwan Department of Physiology, National Cheng Kung University, Tainan 70101, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biochemical Sciences, National Taiwan University, Taipei 10617, Taiwan Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
45
|
Computational analysis of image-based drug profiling predicts synergistic drug combinations: applications in triple-negative breast cancer. Mol Oncol 2014; 8:1548-60. [PMID: 24997502 DOI: 10.1016/j.molonc.2014.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/10/2014] [Indexed: 12/31/2022] Open
Abstract
An imaged-based profiling and analysis system was developed to predict clinically effective synergistic drug combinations that could accelerate the identification of effective multi-drug therapies for the treatment of triple-negative breast cancer and other challenging malignancies. The identification of effective drug combinations for the treatment of triple-negative breast cancer (TNBC) was achieved by integrating high-content screening, computational analysis, and experimental biology. The approach was based on altered cellular phenotypes induced by 55 FDA-approved drugs and biologically active compounds, acquired using fluorescence microscopy and retained in multivariate compound profiles. Dissimilarities between compound profiles guided the identification of 5 combinations, which were assessed for qualitative interaction on TNBC cell growth. The combination of the microtubule-targeting drug vinblastine with KSP/Eg5 motor protein inhibitors monastrol or ispinesib showed potent synergism in 3 independent TNBC cell lines, which was not substantiated in normal fibroblasts. The synergistic interaction was mediated by an increase in mitotic arrest with cells demonstrating typical ispinesib-induced monopolar mitotic spindles, which translated into enhanced apoptosis induction. The antitumour activity of the combination vinblastine/ispinesib was confirmed in an orthotopic mouse model of TNBC. Compared to single drug treatment, combination treatment significantly reduced tumour growth without causing increased toxicity. Image-based profiling and analysis led to the rapid discovery of a drug combination effective against TNBC in vitro and in vivo, and has the potential to lead to the development of new therapeutic options in other hard-to-treat cancers.
Collapse
|
46
|
Abstract
Increased endothelial permeability and reduction of alveolar liquid clearance capacity are two leading pathogenic mechanisms of pulmonary edema, which is a major complication of acute lung injury, severe pneumonia, and acute respiratory distress syndrome, the pathologies characterized by unacceptably high rates of morbidity and mortality. Besides the success in protective ventilation strategies, no efficient pharmacological approaches exist to treat this devastating condition. Understanding of fundamental mechanisms involved in regulation of endothelial permeability is essential for development of barrier protective therapeutic strategies. Ongoing studies characterized specific barrier protective mechanisms and identified intracellular targets directly involved in regulation of endothelial permeability. Growing evidence suggests that, although each protective agonist triggers a unique pattern of signaling pathways, selected common mechanisms contributing to endothelial barrier protection may be shared by different barrier protective agents. Therefore, understanding of basic barrier protective mechanisms in pulmonary endothelium is essential for selection of optimal treatment of pulmonary edema of different etiology. This article focuses on mechanisms of lung vascular permeability, reviews major intracellular signaling cascades involved in endothelial monolayer barrier preservation and summarizes a current knowledge regarding recently identified compounds which either reduce pulmonary endothelial barrier disruption and hyperpermeability, or reverse preexisting lung vascular barrier compromise induced by pathologic insults.
Collapse
Affiliation(s)
- Konstantin G Birukov
- Lung Injury Center, Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA.
| | | | | |
Collapse
|
47
|
Akhshi TK, Wernike D, Piekny A. Microtubules and actin crosstalk in cell migration and division. Cytoskeleton (Hoboken) 2013; 71:1-23. [DOI: 10.1002/cm.21150] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 10/02/2013] [Accepted: 10/06/2013] [Indexed: 12/22/2022]
Affiliation(s)
| | - Denise Wernike
- Department of Biology; Concordia University; Montreal Quebec Canada
| | - Alisa Piekny
- Department of Biology; Concordia University; Montreal Quebec Canada
| |
Collapse
|
48
|
Wang R, Bi J, Ampah KK, Ba X, Liu W, Zeng X. Lipid rafts control human melanoma cell migration by regulating focal adhesion disassembly. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3195-3205. [PMID: 24055995 DOI: 10.1016/j.bbamcr.2013.09.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 08/25/2013] [Accepted: 09/09/2013] [Indexed: 10/26/2022]
Abstract
Tumor cell migration is a crucial step in the metastatic cascade, and interruption of this step is considered to be logically effective in preventing tumor metastasis. Lipid rafts, distinct liquid ordered plasma membrane microdomains, have been shown to influence cancer cell migration, but the underlying mechanisms are still not well understood. Here, we report that lipid rafts regulate the dynamics of actin cytoskeleton and focal adhesion in human melanoma cell migration. Disrupting the integrity of lipid rafts with methyl-β cyclodextrin enhances actin stress fiber formation and inhibits focal adhesion disassembly, accompanied with alterations in cell morphology. Furthermore, actin cytoskeleton, rather than microtubules, mediates the lipid raft-dependent focal adhesion disassembly by regulating the dephosphorylation of focal adhesion proteins and the internalization of β3 integrin. We also show that Src-RhoA-Rho kinase signaling pathway is responsible for lipid raft disruption-induced stress fiber formation. Taken together, these observations provide a new mechanism to further explain how lipid rafts regulate the migration of melanoma cell and suggest that lipid rafts may be novel and attractive targets for cancer therapy.
Collapse
Affiliation(s)
- Ruifei Wang
- Institute of Genetics and Cytology, School of Life Sciences, Northeast Normal University, #5268, Renmin Street, Changchun, Jilin 130024, China
| | - Jiajia Bi
- Institute of Genetics and Cytology, School of Life Sciences, Northeast Normal University, #5268, Renmin Street, Changchun, Jilin 130024, China
| | - Khamal Kwesi Ampah
- Institute of Genetics and Cytology, School of Life Sciences, Northeast Normal University, #5268, Renmin Street, Changchun, Jilin 130024, China
| | - Xueqing Ba
- Institute of Genetics and Cytology, School of Life Sciences, Northeast Normal University, #5268, Renmin Street, Changchun, Jilin 130024, China.
| | - Wenguang Liu
- Institute of Genetics and Cytology, School of Life Sciences, Northeast Normal University, #5268, Renmin Street, Changchun, Jilin 130024, China
| | - Xianlu Zeng
- Institute of Genetics and Cytology, School of Life Sciences, Northeast Normal University, #5268, Renmin Street, Changchun, Jilin 130024, China.
| |
Collapse
|
49
|
Imbalzano KM, Cohet N, Wu Q, Underwood JM, Imbalzano AN, Nickerson JA. Nuclear shape changes are induced by knockdown of the SWI/SNF ATPase BRG1 and are independent of cytoskeletal connections. PLoS One 2013; 8:e55628. [PMID: 23405182 PMCID: PMC3566038 DOI: 10.1371/journal.pone.0055628] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 01/02/2013] [Indexed: 11/24/2022] Open
Abstract
Changes in nuclear morphology occur during normal development and have been observed during the progression of several diseases. The shape of a nucleus is governed by the balance of forces exerted by nuclear-cytoskeletal contacts and internal forces created by the structure of the chromatin and nuclear envelope. However, factors that regulate the balance of these forces and determine nuclear shape are poorly understood. The SWI/SNF chromatin remodeling enzyme ATPase, BRG1, has been shown to contribute to the regulation of overall cell size and shape. Here we document that immortalized mammary epithelial cells show BRG1-dependent nuclear shape changes. Specifically, knockdown of BRG1 induced grooves in the nuclear periphery that could be documented by cytological and ultrastructural methods. To test the hypothesis that the observed changes in nuclear morphology resulted from altered tension exerted by the cytoskeleton, we disrupted the major cytoskeletal networks and quantified the frequency of BRG1-dependent changes in nuclear morphology. The results demonstrated that disruption of cytoskeletal networks did not change the frequency of BRG1-induced nuclear shape changes. These findings suggest that BRG1 mediates control of nuclear shape by internal nuclear mechanisms that likely control chromatin dynamics.
Collapse
Affiliation(s)
- Karen M Imbalzano
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | | | | | | | | | | |
Collapse
|
50
|
Tervaniemi MH, Siitonen HA, Söderhäll C, Minhas G, Vuola J, Tiala I, Sormunen R, Samuelsson L, Suomela S, Kere J, Elomaa O. Centrosomal localization of the psoriasis candidate gene product, CCHCR1, supports a role in cytoskeletal organization. PLoS One 2012. [PMID: 23189171 PMCID: PMC3506594 DOI: 10.1371/journal.pone.0049920] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
CCHCR1 (Coiled-Coil α-Helical Rod protein 1), within the major psoriasis susceptibility locus PSORS1, is a plausible candidate gene with the psoriasis associated risk allele CCHCR1*WWCC. Although its expression pattern in psoriatic skin differs from healthy skin and its overexpression influences cell proliferation in transgenic mice, its role as a psoriasis effector gene has remained unsettled. The 5′-region of the gene contains a SNP (rs3130453) that controls a 5′-extended open reading frame and thus the translation of alternative isoforms. We have now compared the function of two CCHCR1 isoforms: the novel longer isoform 1 and the previously studied isoform 3. In samples of Finnish and Swedish families, the allele generating only isoform 3 shows association with psoriasis (P<10−7). Both isoforms localize at the centrosome, a cell organelle playing a role in cell division. In stably transfected cells the isoform 3 affects cell proliferation and with the CCHCR1*WWCC allele, also apoptosis. Furthermore, cells overexpressing CCHCR1 show isoform- and haplotype-specific influences in the cell size and shape and alterations in the organization and expression of the cytoskeletal proteins actin, vimentin, and cytokeratins. The isoform 1 with the non-risk allele induces the expression of keratin 17, a hallmark for psoriasis; the silencing of CCHCR1 reduces its expression in HEK293 cells. CCHCR1 also regulates EGF-induced STAT3 activation in an isoform-specific manner: the tyrosine phosphorylation of STAT3 is disturbed in isoform 3-transfected cells. The centrosomal localization of CCHCR1 provides a connection to the abnormal cell proliferation and offers a link to possible cellular pathways altered in psoriasis.
Collapse
Affiliation(s)
- Mari H. Tervaniemi
- Haartman Institute, Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Research Program's Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - H. Annika Siitonen
- Haartman Institute, Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Research Program's Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Cilla Söderhäll
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Gurinder Minhas
- Haartman Institute, Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Research Program's Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Jyrki Vuola
- Helsinki Burn Centre, Department of Plastic Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Inkeri Tiala
- Haartman Institute, Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Research Program's Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| | - Raija Sormunen
- Biocenter Oulu, Department of Pathology, University of Oulu, Oulu, Finland
| | - Lena Samuelsson
- Department of Clinical Genetics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sari Suomela
- Department of Dermatology, University of Helsinki, and Helsinki University Central Hospital, Helsinki, Finland
| | - Juha Kere
- Haartman Institute, Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Research Program's Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Outi Elomaa
- Haartman Institute, Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Research Program's Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Folkhälsan Institute of Genetics, Helsinki, Finland
| |
Collapse
|