1
|
Pérez-Sala D, Zorrilla S. Versatility of vimentin assemblies: From filaments to biomolecular condensates and back. Eur J Cell Biol 2025; 104:151487. [PMID: 40194320 DOI: 10.1016/j.ejcb.2025.151487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/26/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025] Open
Abstract
Cytoskeletal structures shape and confer resistance to cells. The intermediate filament protein vimentin forms versatile structures that play key roles in cytoskeletal crosstalk, in the integration of cellular responses to a variety of external and internal cues, and in the defense against stress. Such multifaceted roles can be fulfilled thanks to the vast variety of vimentin proteoforms, which in turn arise from the combinations of a myriad of tightly regulated posttranslational modifications. Diverse vimentin proteoforms will differentially shape its polymeric assemblies, underlying vimentin ability to organize in filaments, bundles, squiggles, droplets, cell surface-bound and/or various secreted forms. Interestingly, certain vimentin dots or droplets have been lately categorized as biomolecular condensates. Biomolecular condensates are phase-separated membraneless structures that are critical for the organization of cellular components and play important roles in pathophysiology. Recent findings have unveiled the importance of low complexity sequence domains in vimentin filament assembly. Moreover, several oxidants trigger the transition of vimentin filaments into phase-separated biomolecular condensates, a reversible process that may provide clues on the role of condensates as seeds for filament formation. Revisiting previous results in the light of recent knowledge prompts the hypothesis that vimentin condensates could play a role in traffic of filament precursors, cytoskeletal crosstalk and cellular responses to stress. Deciphering the "vimentin posttranslational modification code", that is, the structure-function relationships of vimentin proteoforms, constitutes a major challenge to understand the regulation of vimentin behavior and its multiple personalities. This will contribute to unveil essential cellular mechanisms and foster novel opportunities for drug discovery.
Collapse
Affiliation(s)
- Dolores Pérez-Sala
- Department of Molecular and Cellular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain.
| | - Silvia Zorrilla
- Department of Molecular and Cellular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, Madrid, Spain
| |
Collapse
|
2
|
Basu A, Krug T, du Pont B, Huang Q, Sun S, Adam SA, Goldman RD, Weitz DA. Vimentin undergoes liquid-liquid phase separation to form droplets which wet and stabilize actin fibers. Proc Natl Acad Sci U S A 2025; 122:e2418624122. [PMID: 40030010 PMCID: PMC11912372 DOI: 10.1073/pnas.2418624122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025] Open
Abstract
The cytoskeleton is composed of F-actin, microtubules, and intermediate filaments (IFs). Vimentin is one of the most ubiquitous and well-studied IFs. It is involved in many activities including wound healing, tissue fibrosis, and cancer metastasis, all of which require rapid vimentin IF assembly. In this paper, we report that vimentin forms liquid condensates which appear to enable rapid filament growth. Given the transient nature of these droplets, we focus on properties of vimentin-Y117L, which has a point mutation that leads to formation of condensates but not IFs, enabling us to study these droplets in detail. The droplets dissolve under 1,6-Hexanediol treatment and under decreasing concentration, confirming that they are liquid, and phase separated. These condensates extensively wet actin stress fibers, rendering them resistant to actin-binding drugs and protecting them from depolymerization. We show similar behavior occurs in wild-type vimentin during its assembly into filaments.
Collapse
Affiliation(s)
- Arkaprabha Basu
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Tommy Krug
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Benjamin du Pont
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Qiaoling Huang
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Research Institute for Biomimetics and Soft Matter, Fujian Provincial Key Laboratory for Soft Functional Materials Research, Department of Physics, College of Physical Science and Technology, Xiamen University, Xiamen361005, China
| | - Sijie Sun
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
| | - Stephen A. Adam
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Robert D. Goldman
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - David A. Weitz
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA02115
- Department of Physics, Harvard University, Cambridge, MA02138
| |
Collapse
|
3
|
Martínez-Cenalmor P, Martínez AE, Moneo-Corcuera D, González-Jiménez P, Pérez-Sala D. Oxidative stress elicits the remodeling of vimentin filaments into biomolecular condensates. Redox Biol 2024; 75:103282. [PMID: 39079387 PMCID: PMC11338992 DOI: 10.1016/j.redox.2024.103282] [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: 07/02/2024] [Revised: 07/21/2024] [Accepted: 07/21/2024] [Indexed: 08/23/2024] Open
Abstract
The intermediate filament protein vimentin performs an essential role in cytoskeletal interplay and dynamics, mechanosensing and cellular stress responses. In pathology, vimentin is a key player in tumorigenesis, fibrosis and infection. Vimentin filaments undergo distinct and versatile reorganizations, and behave as redox sensors. The vimentin monomer possesses a central α-helical rod domain flanked by N- and C-terminal low complexity domains. Interactions between this type of domains play an important function in the formation of phase-separated biomolecular condensates, which in turn are critical for the organization of cellular components. Here we show that several oxidants, including hydrogen peroxide and diamide, elicit the remodeling of vimentin filaments into small particles. Oxidative stress elicited by diamide induces a fast dissociation of filaments into circular, motile dots, which requires the presence of the single vimentin cysteine residue, C328. This effect is reversible, and filament reassembly can occur within minutes of oxidant removal. Diamide-elicited vimentin droplets recover fluorescence after photobleaching. Moreover, fusion of cells expressing differentially tagged vimentin allows the detection of dots positive for both tags, indicating that vimentin dots merge upon cell fusion. The aliphatic alcohol 1,6-hexanediol, known to alter interactions between low complexity domains, readily dissolves diamide-elicited vimentin dots at low concentrations, in a C328 dependent manner, and hampers reassembly. Taken together, these results indicate that vimentin oxidation promotes a fast and reversible filament remodeling into biomolecular condensate-like structures, and provide primary evidence of its regulated phase separation. Moreover, we hypothesize that filament to droplet transition could play a protective role against irreversible damage of the vimentin network by oxidative stress.
Collapse
Affiliation(s)
- Paula Martínez-Cenalmor
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain
| | - Alma E Martínez
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain
| | - Diego Moneo-Corcuera
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain
| | - Patricia González-Jiménez
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Cellular and Molecular Biosciences, Centro de Investigaciones Biológicas Margarita Salas, CSIC, 28040, Madrid, Spain.
| |
Collapse
|
4
|
Ho Thanh MT, Poudel A, Ameen S, Carroll B, Wu M, Soman P, Zhang T, Schwarz JM, Patteson AE. Vimentin promotes collective cell migration through collagen networks via increased matrix remodeling and spheroid fluidity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599259. [PMID: 38948855 PMCID: PMC11212918 DOI: 10.1101/2024.06.17.599259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The intermediate filament (IF) protein vimentin is associated with many diseases with phenotypes of enhanced cellular migration and aggressive invasion through the extracellular matrix (ECM) of tissues, but vimentin's role in in-vivo cell migration is still largely unclear. Vimentin is important for proper cellular adhesion and force generation, which are critical to cell migration; yet the vimentin cytoskeleton also hinders the ability of cells to squeeze through small pores in ECM, resisting migration. To identify the role of vimentin in collective cell migration, we generate spheroids of wide-type and vimentin-null mouse embryonic fibroblasts (mEFs) and embed them in a 3D collagen matrix. We find that loss of vimentin significantly impairs the ability of the spheroid to collectively expand through collagen networks and remodel the collagen network. Traction force analysis reveals that vimentin null spheroids exert less contractile force than their wild-type counterparts. In addition, spheroids made of mEFs with only vimentin unit length filaments (ULFs) exhibit similar behavior as vimentin-null spheroids, suggesting filamentous vimentin is required to promote 3D collective cell migration. We find the vimentin-mediated collective cell expansion is dependent on matrix metalloproteinase (MMP) degradation of the collagen matrix. Further, 3D vertex model simulation of spheroid and embedded ECM indicates that wild-type spheroids behave more fluid-like, enabling more active pulling and reconstructing the surrounding collagen network. Altogether, these results signify that VIF plays a critical role in enhancing migratory persistence in 3D matrix environments through MMP transportation and tissue fluidity.
Collapse
Affiliation(s)
- Minh Tri Ho Thanh
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - Arun Poudel
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Biomedical and Chemical Engineering Department, Syracuse University; Syracuse, New York, USA
| | - Shabeeb Ameen
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - Bobby Carroll
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - M Wu
- Department of Biological and Environmental Engineering, Cornell University; Ithaca, New York, USA
| | - Pranav Soman
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Biomedical and Chemical Engineering Department, Syracuse University; Syracuse, New York, USA
| | - Tao Zhang
- Department of Polymer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - J M Schwarz
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Indian Creek Farm, Ithaca, New York, USA
| | - Alison E Patteson
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| |
Collapse
|
5
|
Eibauer M, Weber MS, Kronenberg-Tenga R, Beales CT, Boujemaa-Paterski R, Turgay Y, Sivagurunathan S, Kraxner J, Köster S, Goldman RD, Medalia O. Vimentin filaments integrate low-complexity domains in a complex helical structure. Nat Struct Mol Biol 2024; 31:939-949. [PMID: 38632361 PMCID: PMC11189308 DOI: 10.1038/s41594-024-01261-2] [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: 05/08/2023] [Accepted: 03/01/2024] [Indexed: 04/19/2024]
Abstract
Intermediate filaments (IFs) are integral components of the cytoskeleton. They provide cells with tissue-specific mechanical properties and are involved in numerous cellular processes. Due to their intricate architecture, a 3D structure of IFs has remained elusive. Here we use cryo-focused ion-beam milling, cryo-electron microscopy and tomography to obtain a 3D structure of vimentin IFs (VIFs). VIFs assemble into a modular, intertwined and flexible helical structure of 40 α-helices in cross-section, organized into five protofibrils. Surprisingly, the intrinsically disordered head domains form a fiber in the lumen of VIFs, while the intrinsically disordered tails form lateral connections between the protofibrils. Our findings demonstrate how protein domains of low sequence complexity can complement well-folded protein domains to construct a biopolymer with striking mechanical strength and stretchability.
Collapse
Affiliation(s)
- Matthias Eibauer
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| | - Miriam S Weber
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | | - Charlie T Beales
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | | - Yagmur Turgay
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Suganya Sivagurunathan
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Julia Kraxner
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany
- MDC Berlin-Buch, Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany
| | - Robert D Goldman
- Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
6
|
Gomez-Cruz C, Fernandez-de la Torre M, Lachowski D, Prados-de-Haro M, Del Río Hernández AE, Perea G, Muñoz-Barrutia A, Garcia-Gonzalez D. Mechanical and Functional Responses in Astrocytes under Alternating Deformation Modes Using Magneto-Active Substrates. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312497. [PMID: 38610101 DOI: 10.1002/adma.202312497] [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: 11/21/2023] [Revised: 03/21/2024] [Indexed: 04/14/2024]
Abstract
This work introduces NeoMag, a system designed to enhance cell mechanics assays in substrate deformation studies. NeoMag uses multidomain magneto-active materials to mechanically actuate the substrate, transmitting reversible mechanical cues to cells. The system boasts full flexibility in alternating loading substrate deformation modes, seamlessly adapting to both upright and inverted microscopes. The multidomain substrates facilitate mechanobiology assays on 2D and 3D cultures. The integration of the system with nanoindenters allows for precise evaluation of cellular mechanical properties under varying substrate deformation modes. The system is used to study the impact of substrate deformation on astrocytes, simulating mechanical conditions akin to traumatic brain injury and ischemic stroke. The results reveal local heterogeneous changes in astrocyte stiffness, influenced by the orientation of subcellular regions relative to substrate strain. These stiffness variations, exceeding 50% in stiffening and softening, and local deformations significantly alter calcium dynamics. Furthermore, sustained deformations induce actin network reorganization and activate Piezo1 channels, leading to an initial increase followed by a long-term inhibition of calcium events. Conversely, fast and dynamic deformations transiently activate Piezo1 channels and disrupt the actin network, causing long-term cell softening. These findings unveil mechanical and functional alterations in astrocytes during substrate deformation, illustrating the multiple opportunities this technology offers.
Collapse
Affiliation(s)
- Clara Gomez-Cruz
- Department of Continuum Mechanics and Structural Analysis, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911, Leganés, Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911, Leganés, Madrid, Spain
| | - Miguel Fernandez-de la Torre
- Department of Continuum Mechanics and Structural Analysis, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911, Leganés, Madrid, Spain
| | - Dariusz Lachowski
- Department of Continuum Mechanics and Structural Analysis, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911, Leganés, Madrid, Spain
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Martin Prados-de-Haro
- Department of Continuum Mechanics and Structural Analysis, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911, Leganés, Madrid, Spain
| | - Armando E Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK
| | - Gertrudis Perea
- Department of Functional and Systems Neurobiology, Instituto Cajal, CSIC, Av. Doctor Arce, 37., 28002, Leganés, Madrid, Spain
| | - Arrate Muñoz-Barrutia
- Departamento de Bioingeniería, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911, Leganés, Madrid, Spain
- Área de Ingeniería Biomédica, Instituto de Investigación Sanitaria Gregorio Marañón, Calle del Doctor Esquerdo 46, Leganés, Madrid, ES28007, Spain
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, 3400 N Charles St, Baltimore, Maryland, 21218, USA
| | - Daniel Garcia-Gonzalez
- Department of Continuum Mechanics and Structural Analysis, Universidad Carlos III de Madrid, Avda. de la Universidad 30, 28911, Leganés, Madrid, Spain
| |
Collapse
|
7
|
Conboy JP, Istúriz Petitjean I, van der Net A, Koenderink GH. How cytoskeletal crosstalk makes cells move: Bridging cell-free and cell studies. BIOPHYSICS REVIEWS 2024; 5:021307. [PMID: 38840976 PMCID: PMC11151447 DOI: 10.1063/5.0198119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Cell migration is a fundamental process for life and is highly dependent on the dynamical and mechanical properties of the cytoskeleton. Intensive physical and biochemical crosstalk among actin, microtubules, and intermediate filaments ensures their coordination to facilitate and enable migration. In this review, we discuss the different mechanical aspects that govern cell migration and provide, for each mechanical aspect, a novel perspective by juxtaposing two complementary approaches to the biophysical study of cytoskeletal crosstalk: live-cell studies (often referred to as top-down studies) and cell-free studies (often referred to as bottom-up studies). We summarize the main findings from both experimental approaches, and we provide our perspective on bridging the two perspectives to address the open questions of how cytoskeletal crosstalk governs cell migration and makes cells move.
Collapse
Affiliation(s)
- James P. Conboy
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Irene Istúriz Petitjean
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Anouk van der Net
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Gijsje H. Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| |
Collapse
|
8
|
Xie S, Yang X, Yang X, Cao Z, Wei N, Lin X, Shi M, Cao R. Japanese encephalitis virus NS1 and NS1' proteins induce vimentin rearrangement via the CDK1-PLK1 axis to promote viral replication. J Virol 2024; 98:e0019524. [PMID: 38656209 PMCID: PMC11092344 DOI: 10.1128/jvi.00195-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/17/2024] [Indexed: 04/26/2024] Open
Abstract
The host cytoskeleton plays crucial roles in various stages of virus infection, including viral entry, transport, replication, and release. However, the specific mechanisms by which intermediate filaments are involved in orthoflavivirus infection have not been well understood. In this study, we demonstrate that the Japanese encephalitis virus (JEV) remodels the vimentin network, resulting in the formation of cage-like structures that support viral replication. Mechanistically, JEV NS1 and NS1' proteins induce the translocation of CDK1 from the nucleus to the cytoplasm and interact with it, leading to the phosphorylation of vimentin at Ser56. This phosphorylation event recruits PLK1, which further phosphorylates vimentin at Ser83. Consequently, these phosphorylation modifications convert the typically filamentous vimentin into non-filamentous "particles" or "squiggles." These vimentin "particles" or "squiggles" are then transported retrogradely along microtubules to the endoplasmic reticulum, where they form cage-like structures. Notably, NS1' is more effective than NS1 in triggering the CDK1-PLK1 cascade response. Overall, our study provides new insights into how JEV NS1 and NS1' proteins manipulate the vimentin network to facilitate efficient viral replication. IMPORTANCE Japanese encephalitis virus (JEV) is a mosquito-borne orthoflavivirus that causes severe encephalitis in humans, particularly in Asia. Despite the availability of a safe and effective vaccine, JEV infection remains a significant public health threat due to limited vaccination coverage. Understanding the interactions between JEV and host proteins is essential for developing more effective antiviral strategies. In this study, we investigated the role of vimentin, an intermediate filament protein, in JEV replication. Our findings reveal that JEV NS1 and NS1' proteins induce vimentin rearrangement, resulting in the formation of cage-like structures that envelop the viral replication factories (RFs), thus facilitating efficient viral replication. Our research highlights the importance of the interplay between the cytoskeleton and orthoflavivirus, suggesting that targeting vimentin could be a promising approach for the development of antiviral strategies to inhibit JEV propagation.
Collapse
Affiliation(s)
- Shengda Xie
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xiaoxiao Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xingmiao Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ziyu Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ning Wei
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xinxin Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Miaolei Shi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ruibing Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
9
|
Buchwalter A. Intermediate, but not average: The unusual lives of the nuclear lamin proteins. Curr Opin Cell Biol 2023; 84:102220. [PMID: 37619289 PMCID: PMC12049094 DOI: 10.1016/j.ceb.2023.102220] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023]
Abstract
The nuclear lamins are polymeric intermediate filament proteins that scaffold the nucleus and organize the genome in nearly all eukaryotic cells. This review focuses on the dynamic regulation of lamin filaments through their biogenesis, assembly, disassembly, and degradation. The lamins are unusually long-lived proteins under homeostatic conditions, but their turnover can be induced in select contexts that are highlighted in this review. Finally, we discuss recent investigations into the influence of laminopathy-linked mutations on the assembly, folding, and stability of the nuclear lamins.
Collapse
Affiliation(s)
- Abigail Buchwalter
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
10
|
Wang YJ, Liang H, Liu Y, Bao Q, Yang S, Xu XX, Chen YC, Liu W, Shi X, Shi Y, Liu X, Liu B, Gao H, Jiu Y, Liu YJ. Lamin A/C and Vimentin as a Coordinated Regulator during Amoeboid Migration in Microscale Confined Microenvironments. NANO LETTERS 2023; 23:6727-6735. [PMID: 37459599 DOI: 10.1021/acs.nanolett.3c02096] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Cell migration occurs in confined microenvironments, which plays a vital role in the process of tumor metastasis. However, it is challenging to study their behaviors in vivo. Here we developed a cell squeeze system that can be scaled down to micrometers to mimic native physical confined microenvironments, wherein degrees of surface adhesion and mechanical constraints could be manipulated in order to investigate cell-migrating behaviors. Based on the microscale cell squeeze system, we found the synergistic role of lamin A/C and vimentin in cell transition and migration under strong confinement. The dynamic variations in lamin A/C and vimentin expression establish a positive feedback loop in response to confinement, effectively promoting amoeboid migration by modulating nuclear deformability while ensuring cell viability. This work shed light on modulating cell response to microenvironments by altering the expression of lamin A/C and/or vimentin, which may be a more efficient way of inhibiting cancer metastasis.
Collapse
Affiliation(s)
- Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Hong Liang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Yixin Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Qiyuan Bao
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Shanghai 200025, China
| | - Shuang Yang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Xin-Xin Xu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Yu-Chen Chen
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Wei Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Xuemeng Shi
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuheng Shi
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Xiaohui Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Baohong Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Hai Gao
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| | - Yaming Jiu
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Shanghai Stomatological Hospital, Institutes of Biomedical Sciences, Department of Chemistry, Fudan University, Shanghai 200032, China
| |
Collapse
|
11
|
Berr AL, Wiese K, Dos Santos G, Koch CM, Anekalla KR, Kidd M, Davis JM, Cheng Y, Hu YS, Ridge KM. Vimentin is required for tumor progression and metastasis in a mouse model of non-small cell lung cancer. Oncogene 2023:10.1038/s41388-023-02703-9. [PMID: 37161053 DOI: 10.1038/s41388-023-02703-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 11/15/2022] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
Vimentin is highly expressed in metastatic cancers, and its expression correlates with poor patient prognoses. However, no causal in vivo studies linking vimentin and non-small cell lung cancer (NSCLC) progression existed until now. We use three complementary in vivo models to show that vimentin is required for the progression of NSCLC. First, we crossed LSL-KrasG12D; Tp53fl/fl mice (KPV+/+) with vimentin knockout mice (KPV-/-) to demonstrate that KPV-/- mice have attenuated tumor growth and improved survival compared with KPV+/+ mice. Next, we therapeutically treated KPV+/+ mice with withaferin A (WFA), an agent that disrupts vimentin intermediate filaments (IFs). We show that WFA suppresses tumor growth and reduces tumor burden in the lung. Finally, luciferase-expressing KPV+/+, KPV-/-, or KPVY117L cells were implanted into the flanks of athymic mice to track cancer metastasis to the lung. In KPVY117L cells, vimentin forms oligomers called unit-length filaments but cannot assemble into mature vimentin IFs. KPV-/- and KPVY117L cells fail to metastasize, suggesting that cell-autonomous metastasis requires mature vimentin IFs. Integrative metabolomic and transcriptomic analysis reveals that KPV-/- cells upregulate genes associated with ferroptosis, an iron-dependent form of regulated cell death. KPV-/- cells have reduced glutathione peroxidase 4 (GPX4) levels, resulting in the accumulation of toxic lipid peroxides and increased ferroptosis. Together, our results demonstrate that vimentin is required for rapid tumor growth, metastasis, and protection from ferroptosis in NSCLC.
Collapse
Affiliation(s)
- Alexandra L Berr
- Department of Biomedical Engineering, Northwestern University, Chicago, IL, USA
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Kristin Wiese
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Gimena Dos Santos
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Clarissa M Koch
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Kishore R Anekalla
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Martha Kidd
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Jennifer M Davis
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Yuan Cheng
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Yuan-Shih Hu
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA.
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
12
|
Renganathan B, Zewe JP, Cheng Y, Paumier J, Kittisopikul M, Ridge KM, Opal P, Gelfand VI. Gigaxonin is required for intermediate filament transport. FASEB J 2023; 37:e22886. [PMID: 37043392 PMCID: PMC10237250 DOI: 10.1096/fj.202202119r] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/23/2023] [Accepted: 03/08/2023] [Indexed: 04/13/2023]
Abstract
Gigaxonin is an adaptor protein for E3 ubiquitin ligase substrates. It is necessary for ubiquitination and degradation of intermediate filament (IF) proteins. Giant axonal neuropathy is a pathological condition caused by mutations in the GAN gene that encodes gigaxonin. This condition is characterized by abnormal accumulation of IFs in both neuronal and non-neuronal cells; however, it is unclear what causes IF aggregation. In this work, we studied the dynamics of IFs using their subunits tagged with a photoconvertible protein mEOS 3.2. We have demonstrated that the loss of gigaxonin dramatically inhibited transport of IFs along microtubules by the microtubule motor kinesin-1. This inhibition was specific for IFs, as other kinesin-1 cargoes, with the exception of mitochondria, were transported normally. Abnormal distribution of IFs in the cytoplasm can be rescued by direct binding of kinesin-1 to IFs, demonstrating that transport inhibition is the primary cause for the abnormal IF distribution. Another effect of gigaxonin loss was a more than 20-fold increase in the amount of soluble vimentin oligomers in the cytosol of gigaxonin knock-out cells. We speculate that these oligomers saturate a yet unidentified adapter that is required for kinesin-1 binding to IFs, which might inhibit IF transport along microtubules causing their abnormal accumulation.
Collapse
Affiliation(s)
- Bhuvanasundar Renganathan
- Department of Cell and Developmental BiologyFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - James P. Zewe
- Ken and Ruth Davee Department of NeurologyFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Yuan Cheng
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineNorthwestern University, Feinberg School of MedicineChicagoIllinoisUSA
| | - Jean‐Michel Paumier
- Ken and Ruth Davee Department of NeurologyFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Mark Kittisopikul
- Department of Cell and Developmental BiologyFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Karen M. Ridge
- Division of Pulmonary and Critical Care MedicineDepartment of MedicineNorthwestern University, Feinberg School of MedicineChicagoIllinoisUSA
| | - Puneet Opal
- Ken and Ruth Davee Department of NeurologyFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | - Vladimir I. Gelfand
- Department of Cell and Developmental BiologyFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
13
|
Lee YA, Lek CCJ, Rong G, Wu Z, Shathishwaran S, Lee JHJ, Tam WL, Wuestefeld T, Park SJ, Jung S, Kim B, Kang NY, Chang YT. Theranostics application of tumor-initiating cell probe TiY in non-small cell lung cancer. Theranostics 2023; 13:1370-1380. [PMID: 36923526 PMCID: PMC10008731 DOI: 10.7150/thno.79282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/19/2023] [Indexed: 03/13/2023] Open
Abstract
Background: Tumor-initiating cells (TIC) often elude conventional cancer treatment, which results in metastasis and cancer relapse. Recently, studies have begun to focus on the TIC population in tumors to provide better therapeutic options. Previously, we have reported the successful development of a TIC-specific probe TiY with the binding target as vimentin. While a low concentration of TiY showed a TIC visualization, at a high concentration, TiY induced selective toxicity onto TIC in vitro. In this study, we aim to assess TiY's applicability in theranostics purposes, from in vivo visualization to therapeutic effect toward TIC, in cancer mouse models. Methods: We performed cell experiments with the TIC line model derived from resected primary non-small cell lung cancer (NSCLC) patient tumor. The animal model studies were conducted in mice of NSCLC patient-derived xenograft (PDX). TiY was intravenously delivered into the mice models at different concentrations to assess its in vivo TIC-selective staining and therapeutic effect. Results: We demonstrated the TIC-selective identification and therapeutic effect of TiY in animal models. TiY treatment induced a significant ablation of the TIC population in the tumor, and further molecular study elucidated that the mechanism of TiY is through vimentin dynamics in TIC. Conclusion: The results underscore the applicability of TiY for cancer treatment by selectively targeting soluble vimentin in TIC.
Collapse
Affiliation(s)
- Yong-An Lee
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (ASTAR), Singapore, 138672, Singapore
| | - Chee Chong Jonathan Lek
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (ASTAR), Singapore, 138672, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Gao Rong
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (ASTAR), Singapore, 138672, Singapore
| | - Zhengwei Wu
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (ASTAR), Singapore, 138672, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - S Shathishwaran
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (ASTAR), Singapore, 138672, Singapore
| | - Jia Hui Jane Lee
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (ASTAR), Singapore, 138672, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (ASTAR), Singapore, 138672, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Torsten Wuestefeld
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (ASTAR), Singapore, 138672, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore.,National Cancer Centre Singapore, 169610, Singapore
| | - Sung-Jin Park
- Institute of Bioengineering and Bioimaging (IBB), Agency for Science, Technology and Research (ASTAR), Singapore, 138667, Singapore
| | - Sangyong Jung
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (ASTAR), Singapore, 138673, Singapore
| | - Beomsue Kim
- Neural Circuit Research Group, Korea Brain Research Institute (KBRI) Daegu, 41068, Republic of Korea
| | - Nam-Young Kang
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea
| | - Young-Tae Chang
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS), Pohang, 37673, Republic of Korea.,Department of Chemistry, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Republic of Korea
| |
Collapse
|
14
|
Ndiaye AB, Koenderink GH, Shemesh M. Intermediate Filaments in Cellular Mechanoresponsiveness: Mediating Cytoskeletal Crosstalk From Membrane to Nucleus and Back. Front Cell Dev Biol 2022; 10:882037. [PMID: 35478961 PMCID: PMC9035595 DOI: 10.3389/fcell.2022.882037] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/24/2022] [Indexed: 11/25/2022] Open
Abstract
The mammalian cytoskeleton forms a mechanical continuum that spans across the cell, connecting the cell surface to the nucleus via transmembrane protein complexes in the plasma and nuclear membranes. It transmits extracellular forces to the cell interior, providing mechanical cues that influence cellular decisions, but also actively generates intracellular forces, enabling the cell to probe and remodel its tissue microenvironment. Cells adapt their gene expression profile and morphology to external cues provided by the matrix and adjacent cells as well as to cell-intrinsic changes in cytoplasmic and nuclear volume. The cytoskeleton is a complex filamentous network of three interpenetrating structural proteins: actin, microtubules, and intermediate filaments. Traditionally the actin cytoskeleton is considered the main contributor to mechanosensitivity. This view is now shifting owing to the mounting evidence that the three cytoskeletal filaments have interdependent functions due to cytoskeletal crosstalk, with intermediate filaments taking a central role. In this Mini Review we discuss how cytoskeletal crosstalk confers mechanosensitivity to cells and tissues, with a particular focus on the role of intermediate filaments. We propose a view of the cytoskeleton as a composite structure, in which cytoskeletal crosstalk regulates the local stability and organization of all three filament families at the sub-cellular scale, cytoskeletal mechanics at the cellular scale, and cell adaptation to external cues at the tissue scale.
Collapse
Affiliation(s)
| | | | - Michal Shemesh
- *Correspondence: Michal Shemesh, ; Gijsje H. Koenderink,
| |
Collapse
|
15
|
Ostrowska-Podhorodecka Z, Ding I, Norouzi M, McCulloch CA. Impact of Vimentin on Regulation of Cell Signaling and Matrix Remodeling. Front Cell Dev Biol 2022; 10:869069. [PMID: 35359446 PMCID: PMC8961691 DOI: 10.3389/fcell.2022.869069] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/25/2022] [Indexed: 12/12/2022] Open
Abstract
Vimentin expression contributes to cellular mechanoprotection and is a widely recognized marker of fibroblasts and of epithelial-mesenchymal transition. But it is not understood how vimentin affects signaling that controls cell migration and extracellular matrix (ECM) remodeling. Recent data indicate that vimentin controls collagen deposition and ECM structure by regulating contractile force application to the ECM and through post-transcriptional regulation of ECM related genes. Binding of cells to the ECM promotes the association of vimentin with cytoplasmic domains of adhesion receptors such as integrins. After initial adhesion, cell-generated, myosin-dependent forces and signals that impact vimentin structure can affect cell migration. Post-translational modifications of vimentin determine its adaptor functions, including binding to cell adhesion proteins like paxillin and talin. Accordingly, vimentin regulates the growth, maturation and adhesive strength of integrin-dependent adhesions, which enables cells to tune their attachment to collagen, regulate the formation of cell extensions and control cell migration through connective tissues. Thus, vimentin tunes signaling cascades that regulate cell migration and ECM remodeling. Here we consider how specific properties of vimentin serve to control cell attachment to the underlying ECM and to regulate mesenchymal cell migration and remodeling of the ECM by resident fibroblasts.
Collapse
|
16
|
Nunes Vicente F, Lelek M, Tinevez JY, Tran QD, Pehau-Arnaudet G, Zimmer C, Etienne-Manneville S, Giannone G, Leduc C. Molecular organization and mechanics of single vimentin filaments revealed by super-resolution imaging. SCIENCE ADVANCES 2022; 8:eabm2696. [PMID: 35213220 PMCID: PMC8880768 DOI: 10.1126/sciadv.abm2696] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/11/2022] [Indexed: 05/30/2023]
Abstract
Intermediate filaments (IFs) are involved in key cellular functions including polarization, migration, and protection against large deformations. These functions are related to their remarkable ability to extend without breaking, a capacity that should be determined by the molecular organization of subunits within filaments. However, this structure-mechanics relationship remains poorly understood at the molecular level. Here, using super-resolution microscopy (SRM), we show that vimentin filaments exhibit a ~49-nanometer axial repeat both in cells and in vitro. As unit-length filaments (ULFs) were measured at ~59 nanometers, this demonstrates a partial overlap of ULFs during filament assembly. Using an SRM-compatible stretching device, we also provide evidence that the extensibility of vimentin is due to the unfolding of its subunits and not to their sliding, thus establishing a direct link between the structural organization and its mechanical properties. Overall, our results pave the way for future studies of IF assembly, mechanical, and structural properties in cells.
Collapse
Affiliation(s)
- Filipe Nunes Vicente
- Institut Interdisciplinaire des Neurosciences, CNRS UMR 5297, Université de Bordeaux, Bordeaux F-33000, France
| | - Mickael Lelek
- Imaging and Modeling Unit, Institut Pasteur, CNRS UMR 3691, Paris F-75015, France
| | - Jean-Yves Tinevez
- Image Analysis Hub, 2RT / DTPS, Institut Pasteur, Paris F-75015 , France
| | - Quang D. Tran
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, CNRS UMR 3691, équipe labellisée Ligue contre le cancer, Paris F-75015, France
- CNRS UMR 7592, Institut Jacques Monod, Université de Paris, Paris F-75013, France
| | - Gerard Pehau-Arnaudet
- CNRS UMR 3528, Institut Pasteur, Paris F-75015, France
- Ultrastructural BioImaging Platform, Institut Pasteur, Paris F-75015, France
| | - Christophe Zimmer
- Imaging and Modeling Unit, Institut Pasteur, CNRS UMR 3691, Paris F-75015, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, CNRS UMR 3691, équipe labellisée Ligue contre le cancer, Paris F-75015, France
| | - Gregory Giannone
- Institut Interdisciplinaire des Neurosciences, CNRS UMR 5297, Université de Bordeaux, Bordeaux F-33000, France
| | - Cécile Leduc
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, CNRS UMR 3691, équipe labellisée Ligue contre le cancer, Paris F-75015, France
- CNRS UMR 7592, Institut Jacques Monod, Université de Paris, Paris F-75013, France
| |
Collapse
|
17
|
Vimentin: Regulation and pathogenesis. Biochimie 2022; 197:96-112. [DOI: 10.1016/j.biochi.2022.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 01/11/2022] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
|
18
|
Chu CT, Chen YH, Chiu WT, Chen HC. Tyrosine phosphorylation of lamin A by Src promotes disassembly of nuclear lamina in interphase. Life Sci Alliance 2021; 4:4/10/e202101120. [PMID: 34385357 PMCID: PMC8362257 DOI: 10.26508/lsa.202101120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/01/2021] [Accepted: 08/03/2021] [Indexed: 11/24/2022] Open
Abstract
Lamins form the nuclear lamina, which is important for nuclear structure and activity. Although posttranslational modifications, in particular serine phosphorylation, have been shown to be important for structural properties and functions of lamins, little is known about the role of tyrosine phosphorylation in this regard. In this study, we found that the constitutively active Src Y527F mutant caused the disassembly of lamin A/C. We demonstrate that Src directly phosphorylates lamin A mainly at Tyr45 both in vitro and in intact cells. The phosphomimetic Y45D mutant was diffusively distributed in the nucleoplasm and failed to assemble into the nuclear lamina. Depletion of lamin A/C in HeLa cells induced nuclear dysmorphia and genomic instability as well as increased nuclear plasticity for cell migration, all of which were partially restored by re-expression of lamin A, but further promoted by the Y45D mutant. Together, our results reveal a novel mechanism for regulating the assembly of nuclear lamina through Src and suggest that aberrant phosphorylation of lamin A by Src may contribute to nuclear dysmorphia, genomic instability, and nuclear plasticity.
Collapse
Affiliation(s)
- Ching-Tung Chu
- Institue of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsuan Chen
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Hong-Chen Chen
- Institue of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan .,Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institue of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
19
|
Fan JQ, Li YJ, Wei ZJ, Fan Y, Li XD, Chen ZM, Hou DY, Xiao WY, Ding MR, Wang H, Wang L. Binding-Induced Fibrillogenesis Peptides Recognize and Block Intracellular Vimentin Skeletonization against Breast Cancer. NANO LETTERS 2021; 21:6202-6210. [PMID: 34259530 DOI: 10.1021/acs.nanolett.1c01950] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Life is recognized as a sophisticated self-assembling material system. Cancer involves the overexpression and improper self-assembly of proteins, such as cytoskeleton protein vimentin, an emerging target related to tumor metastasis. Herein, we design a binding-induced fibrillogenesis (BIF) peptide that in situ forms fibrous networks, blocking the improper self-assembly of vimentin against cancer. The BIF peptide can bind to vimentin and subsequently perform fibrillogenesis to form fibers on vimentin. The resultant peptide fibrous network blocks vimentin skeletonization and inhibits the migration and invasion of tumor cells. In mouse models of tumor metastasis, the volume of tumor and the number of lung metastases are markedly decreased. Moreover, the efficacy of BIF peptide (5 mg/kg) is much higher than small molecular antimetastasis drug withaferin A (5 mg/kg) as a standard, indicating that the BIF peptide shows advantages over small molecular inhibitors in blocking the intracellular protein self-assembly.
Collapse
Affiliation(s)
- Jia-Qi Fan
- Hubei Key Laboratory of Catalysis and Materials Science, South-Central University for Nationalities, 182 Minzu Road, Hongshan District, Wuhan, Hubei 430074, People's Republic of China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Yi-Jing Li
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Zi-Jin Wei
- Hubei Key Laboratory of Catalysis and Materials Science, South-Central University for Nationalities, 182 Minzu Road, Hongshan District, Wuhan, Hubei 430074, People's Republic of China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Yu Fan
- Hubei Key Laboratory of Catalysis and Materials Science, South-Central University for Nationalities, 182 Minzu Road, Hongshan District, Wuhan, Hubei 430074, People's Republic of China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Xiang-Dan Li
- Hubei Key Laboratory of Catalysis and Materials Science, South-Central University for Nationalities, 182 Minzu Road, Hongshan District, Wuhan, Hubei 430074, People's Republic of China
| | - Zi-Ming Chen
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Da-Yong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Wu-Yi Xiao
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Meng-Ru Ding
- Hubei Key Laboratory of Catalysis and Materials Science, South-Central University for Nationalities, 182 Minzu Road, Hongshan District, Wuhan, Hubei 430074, People's Republic of China
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, People's Republic of China
| |
Collapse
|
20
|
Abstract
Cell morphology, architecture and dynamics primarily rely on intracellular cytoskeletal networks, which in metazoans are mainly composed of actin microfilaments, microtubules and intermediate filaments (IFs). The diameter size of 10 nm - intermediate between the diameters of actin microfilaments and microtubules - initially gave IFs their name. However, the structure, dynamics, mechanical properties and functions of IFs are not intermediate but set them apart from actin and microtubules. Because of their nucleotide-independent assembly, the lack of intrinsic polarity, their relative stability and their complex composition, IFs had long been overlooked by cell biologists. Now, the numerous human diseases identified to be associated with IF gene mutations and the accumulating evidence of IF functions in cell and tissue integrity explain the growing attention that is being given to the structural characteristics, dynamics and functions of these filaments. In this Primer, we highlight the growing evidence that has revealed a role for IFs as a key element of the cytoskeleton, providing versatile, tunable, cell-type-specific filamentous networks with unique cytoplasmic and nuclear functions.
Collapse
Affiliation(s)
- Gaëlle Dutour-Provenzano
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Équipe Labellisée Ligue Contre le Cancer, F-75015 Paris, France; Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Équipe Labellisée Ligue Contre le Cancer, F-75015 Paris, France.
| |
Collapse
|
21
|
Ostrowska-Podhorodecka Z, McCulloch CA. Vimentin regulates the assembly and function of matrix adhesions. Wound Repair Regen 2021; 29:602-612. [PMID: 33887795 DOI: 10.1111/wrr.12920] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022]
Abstract
The intermediate filament protein vimentin is a widely used phenotypic marker for identifying cells of the mesenchymal linkage such as fibroblasts and myofibroblasts, but the full repertoire of vimentin's functional attributes has not been fully explored. Here we consider how vimentin, in addition to its contributions to mechanical stabilization of cell structure, also helps to control the assembly of cell adhesions and migration through collagen matrices. While the assembly and function of matrix adhesions are critical for the differentiation of myofibroblasts and many other types of adherent cells, a potential mechanism that explains how vimentin affects the recruitment and abundance of centrally important proteins in cell adhesions has been elusive. Here we review recent data indicating that vimentin plays a central regulatory role in the assembly of focal adhesions which form in response to the attachment to collagen. We show that in particular, vimentin is a key organizer of the β1 integrin adhesive machinery, which affects cell migration through collagen. This review provides a comprehensive picture of the surprisingly broad array of processes and molecules with which vimentin interacts to affect cell function in the context of fibroblast and myofibroblast adhesion and migration on collagen.
Collapse
|
22
|
Yamamoto M, Sakamoto Y, Honda Y, Koike K, Nakamura H, Matsumoto T, Ando S. De novo filament formation by human hair keratins K85 and K35 follows a filament development pattern distinct from cytokeratin filament networks. FEBS Open Bio 2021; 11:1299-1312. [PMID: 33605551 PMCID: PMC8091587 DOI: 10.1002/2211-5463.13126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 01/24/2021] [Accepted: 02/18/2021] [Indexed: 11/23/2022] Open
Abstract
In human hair follicles, the hair‐forming cells express 16 hair keratin genes depending on the differentiation stages. K85 and K35 are the first hair keratins expressed in cortical cells at the early stage of the differentiation. Two types of mutations in the gene encoding K85 are associated with ectodermal dysplasia of hair and nail type. Here, we transfected cultured SW‐13 cells with human K85 and K35 genes and characterized filament formation. The K85–K35 pair formed short filaments in the cytoplasm, which gradually elongated and became thicker and entangled around the nucleus, indicating that K85–K35 promotes lateral association of short intermediate filaments (IFs) into bundles but cannot form IF networks in the cytoplasm. Of the K85 mutations related to ectodermal dysplasia of hair and nail type, a two‐nucleotide (C1448T1449) deletion (delCT) in the protein tail domain of K85 interfered with the K85–K35 filament formation and gave only aggregates, whereas a missense mutation (233A>G) that replaces Arg78 with His (R78H) in the head domain of K85 did not interfere with the filament formation. Transfection of cultured MCF‐7 cells with all the hair keratin gene combinations, K85–K35, K85(R78H)–K35 and K85(delCT)–K35, as well as the individual hair keratin genes, formed well‐developed cytoplasmic IF networks, probably by incorporating into the endogenous cytokeratin IF networks. Thus, the unique de novo assembly properties of the K85–K35 pair might play a key role in the early stage of hair formation.
Collapse
Affiliation(s)
- Masaki Yamamoto
- Faculty of Biotechnology and Life Science, Sojo University, Kumamoto, Japan
| | - Yasuko Sakamoto
- Faculty of Biotechnology and Life Science, Sojo University, Kumamoto, Japan
| | - Yuko Honda
- Faculty of Medicine, Saga University, Japan
| | - Kenzo Koike
- Hair Care Research Center, KAO Corporation, Tokyo, Japan
| | - Hideaki Nakamura
- Faculty of Pharmaceutical Science, Sojo University, Kumamoto, Japan
| | | | - Shoji Ando
- Faculty of Biotechnology and Life Science, Sojo University, Kumamoto, Japan
| |
Collapse
|
23
|
Patteson AE, Carroll RJ, Iwamoto DV, Janmey PA. The vimentin cytoskeleton: when polymer physics meets cell biology. Phys Biol 2020; 18:011001. [PMID: 32992303 PMCID: PMC8240483 DOI: 10.1088/1478-3975/abbcc2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The proper functions of tissues depend on the ability of cells to withstand stress and maintain shape. Central to this process is the cytoskeleton, comprised of three polymeric networks: F-actin, microtubules, and intermediate filaments (IFs). IF proteins are among the most abundant cytoskeletal proteins in cells; yet they remain some of the least understood. Their structure and function deviate from those of their cytoskeletal partners, F-actin and microtubules. IF networks show a unique combination of extensibility, flexibility and toughness that confers mechanical resilience to the cell. Vimentin is an IF protein expressed in mesenchymal cells. This review highlights exciting new results on the physical biology of vimentin intermediate filaments and their role in allowing whole cells and tissues to cope with stress.
Collapse
Affiliation(s)
- Alison E Patteson
- Physics Department, Syracuse University, Syracuse, NY 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Robert J Carroll
- Physics Department, Syracuse University, Syracuse, NY 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Daniel V Iwamoto
- Institute for Medicine and Engineering, Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul A Janmey
- Institute for Medicine and Engineering, Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Ding I, Ostrowska-Podhorodecka Z, Lee W, Liu RS, Carneiro K, Janmey PA, McCulloch CA. Cooperative roles of PAK1 and filamin A in regulation of vimentin assembly and cell extension formation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118739. [DOI: 10.1016/j.bbamcr.2020.118739] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/30/2020] [Accepted: 05/05/2020] [Indexed: 01/02/2023]
|
25
|
Bott CJ, Winckler B. Intermediate filaments in developing neurons: Beyond structure. Cytoskeleton (Hoboken) 2020; 77:110-128. [PMID: 31970897 DOI: 10.1002/cm.21597] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 12/20/2022]
Abstract
Neuronal development relies on a highly choreographed progression of dynamic cellular processes by which newborn neurons migrate, extend axons and dendrites, innervate their targets, and make functional synapses. Many of these dynamic processes require coordinated changes in morphology, powered by the cell's cytoskeleton. Intermediate filaments (IFs) are the third major cytoskeletal elements in vertebrate cells, but are rarely considered when it comes to understanding axon and dendrite growth, pathfinding and synapse formation. In this review, we first introduce the many new and exciting concepts of IF function, discovered mostly in non-neuronal cells. These roles include dynamic rearrangements, crosstalk with microtubules and actin filaments, mechano-sensing and -transduction, and regulation of signaling cascades. We then discuss the understudied roles of neuronally expressed IFs, with a particular focus on IFs expressed during development, such as nestin, vimentin and α-internexin. Lastly, we illustrate how signaling modulation by the unconventional IF nestin shapes neuronal morphogenesis in unexpected and novel ways. Even though the first IF knockout mice were made over 20 years ago, the study of the cell biological functions of IFs in the brain still has much room for exciting new discoveries.
Collapse
Affiliation(s)
- Christopher J Bott
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
26
|
Quantitative SUMO proteomics identifies PIAS1 substrates involved in cell migration and motility. Nat Commun 2020; 11:834. [PMID: 32047143 PMCID: PMC7012886 DOI: 10.1038/s41467-020-14581-w] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 01/07/2020] [Indexed: 01/09/2023] Open
Abstract
The protein inhibitor of activated STAT1 (PIAS1) is an E3 SUMO ligase that plays important roles in various cellular pathways. Increasing evidence shows that PIAS1 is overexpressed in various human malignancies, including prostate and lung cancers. Here we used quantitative SUMO proteomics to identify potential substrates of PIAS1 in a system-wide manner. We identified 983 SUMO sites on 544 proteins, of which 62 proteins were assigned as putative PIAS1 substrates. In particular, vimentin (VIM), a type III intermediate filament protein involved in cytoskeleton organization and cell motility, was SUMOylated by PIAS1 at Lys-439 and Lys-445 residues. VIM SUMOylation was necessary for its dynamic disassembly and cells expressing a non-SUMOylatable VIM mutant showed a reduced level of migration. Our approach not only enables the identification of E3 SUMO ligase substrates but also yields valuable biological insights into the unsuspected role of PIAS1 and VIM SUMOylation on cell motility. PIAS1 is an E3 SUMO ligase involved in various cellular processes. Here, the authors use quantitative proteomics to identify potential PIAS1 substrates in human cells and elucidate the biological consequences of PIAS1-mediated SUMOylation of vimentin.
Collapse
|
27
|
Vakhrusheva A, Endzhievskaya S, Zhuikov V, Nekrasova T, Parshina E, Ovsiannikova N, Popov V, Bagrov D, Minin AА, Sokolova OS. The role of vimentin in directional migration of rat fibroblasts. Cytoskeleton (Hoboken) 2019; 76:467-476. [PMID: 31626376 DOI: 10.1002/cm.21572] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 10/05/2019] [Accepted: 10/14/2019] [Indexed: 12/16/2022]
Abstract
Cell migration is one of the most important processes in which the cytoskeleton plays a main role. The cytoskeleton network is formed by tubulin microtubules, actin filaments, and intermediate filaments (IFs). While the structure and functions of the two aforementioned proteins have been extensively investigated during the last decades, vimentin IFs structure and their role in cell migration and adhesion remain unclear. Here, we investigated polarity determination in rat fibroblasts with either a knocked out vim gene or with a mutation that blocks filament formation on the stage of unit-length filaments (ULFs). Structured illumination microscopy has demonstrated the difference in the morphology of IFs in wild-type fibroblasts and of ULFs in mutant fibroblasts. We have developed an approach to measure cell stiffness separately on the trailing and leading edges using atomic force microscopy. Young's modulus values on the leading and trailing edge of migrating rat fibroblasts differ approximately by two times, being larger on the leading edge. The knockout of the vim gene leads to having comparable values of Young's moduli on both edges. Vimentin-null cells change the direction of migration more frequently than those expressing wild-type or mutated vimentin. Our results have shown the principle role of vimentin, not only in the form of IFs, but also as ULFs, in the determination of the polarity and the directionality of fibroblast migration.
Collapse
Affiliation(s)
- Anna Vakhrusheva
- Lomonosov Moscow State University, Department of Biology, Moscow, Russia
| | - Sofia Endzhievskaya
- Institute of Protein Research of Russian Academy of Sciences, Department of Cell Biology, Moscow, Russia
| | - Vsevolod Zhuikov
- Research Centre of Biotechnology of Russian Academy of Sciences, Moscow, Russia
| | - Tatyana Nekrasova
- Institute of Protein Research of Russian Academy of Sciences, Department of Cell Biology, Moscow, Russia
| | - Evgenia Parshina
- Lomonosov Moscow State University, Department of Biology, Moscow, Russia
| | - Natalia Ovsiannikova
- Lomonosov Moscow State University, Belozersky Institute of Physico-chemical biology, Moscow, Russia
| | - Vladimir Popov
- Lomonosov Moscow State University, Department of Physics, Moscow, Russia
| | - Dmitry Bagrov
- Lomonosov Moscow State University, Department of Biology, Moscow, Russia
| | - Alexander А Minin
- Institute of Protein Research of Russian Academy of Sciences, Department of Cell Biology, Moscow, Russia
| | - Olga S Sokolova
- Lomonosov Moscow State University, Department of Biology, Moscow, Russia
| |
Collapse
|
28
|
Duarte S, Viedma-Poyatos Á, Navarro-Carrasco E, Martínez AE, Pajares MA, Pérez-Sala D. Vimentin filaments interact with the actin cortex in mitosis allowing normal cell division. Nat Commun 2019; 10:4200. [PMID: 31519880 PMCID: PMC6744490 DOI: 10.1038/s41467-019-12029-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 08/09/2019] [Indexed: 01/27/2023] Open
Abstract
The vimentin network displays remarkable plasticity to support basic cellular functions and reorganizes during cell division. Here, we show that in several cell types vimentin filaments redistribute to the cell cortex during mitosis, forming a robust framework interwoven with cortical actin and affecting its organization. Importantly, the intrinsically disordered tail domain of vimentin is essential for this redistribution, which allows normal mitotic progression. A tailless vimentin mutant forms curly bundles, which remain entangled with dividing chromosomes leading to mitotic catastrophes or asymmetric partitions. Serial deletions of vimentin tail domain gradually impair cortical association and mitosis progression. Disruption of f-actin, but not of microtubules, causes vimentin bundling near the chromosomes. Pathophysiological stimuli, including HIV-protease and lipoxidation, induce similar alterations. Interestingly, full filament formation is dispensable for cortical association, which also occurs in vimentin particles. These results unveil implications of vimentin dynamics in cell division through its interplay with the actin cortex. The intermediate filament vimentin reorganizes during mitosis, but its molecular regulation and impact on the cell during cell division is unclear. Here, the authors show that vimentin filaments redistribute to the cell cortex during mitosis intertwining with and affecting actin organization.
Collapse
Affiliation(s)
- Sofia Duarte
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Álvaro Viedma-Poyatos
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Elena Navarro-Carrasco
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Alma E Martínez
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain.
| |
Collapse
|
29
|
Terriac E, Schütz S, Lautenschläger F. Vimentin Intermediate Filament Rings Deform the Nucleus During the First Steps of Adhesion. Front Cell Dev Biol 2019; 7:106. [PMID: 31263698 PMCID: PMC6590062 DOI: 10.3389/fcell.2019.00106] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022] Open
Abstract
During cell spreading, cells undergo many changes to their architecture and their mechanical properties. Vimentin, as an integral part of the cell architecture, and its mechanical stability must adapt to the new state of the cell. This study focuses on the structures formed by vimentin during the first steps of cell adhesion. Very early, ball-like structures, or "knots," are seen and often vimentin filaments emerge in the shape of rings around the nucleus. Although intermediate filaments are not known to be associated to motor proteins to form contractile systems, these rings can nonetheless strongly deform the cell nucleus. In the first 6 to 12 h of adhesion, these vimentin knots and rings disappear, and the intermediate filament network returns to the state seen before detachment of the cells. As these vimentin structures are very transient in the early steps of cell spreading, they have rarely been described in the literature. However, they can also be seen during mitosis, which is an event that involves partial detachment and re-spreading of the cells. Interestingly, the turnover dynamics of vimentin are reduced in both the knots and rings, compared to vimentin in the lamellipodia. It remains to define how the force is transmitted from the ball-like structures to the rings, and to measure the impact of such strong nuclear deformation on gene expression during cell re-spreading and the rearrangement of the vimentin network.
Collapse
Affiliation(s)
| | - Susanne Schütz
- Faculty of Natural Sciences and Technology, Saarland University, Saarbrücken, Germany
| | - Franziska Lautenschläger
- Leibniz Institute for New Materials, Saarbrücken, Germany
- Faculty of Natural Sciences and Technology, Saarland University, Saarbrücken, Germany
| |
Collapse
|
30
|
Mónico A, Duarte S, Pajares MA, Pérez-Sala D. Vimentin disruption by lipoxidation and electrophiles: Role of the cysteine residue and filament dynamics. Redox Biol 2019; 23:101098. [PMID: 30658903 PMCID: PMC6859561 DOI: 10.1016/j.redox.2019.101098] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/28/2018] [Accepted: 01/05/2019] [Indexed: 12/17/2022] Open
Abstract
The intermediate filament protein vimentin constitutes a critical sensor for electrophilic and oxidative stress, which induce extensive reorganization of the vimentin cytoskeletal network. Here, we have investigated the mechanisms underlying these effects. In vitro, electrophilic lipids, including 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) and 4-hydroxynonenal (HNE), directly bind to vimentin, whereas the oxidant diamide induces disulfide bond formation. Mutation of the single vimentin cysteine residue (Cys328) blunts disulfide formation and reduces lipoxidation by 15d-PGJ2, but not HNE. Preincubation with these agents differentially hinders NaCl-induced filament formation by wild-type vimentin, with effects ranging from delayed elongation and increased filament diameter to severe impairment of assembly or aggregation. Conversely, the morphology of vimentin Cys328Ser filaments is mildly or not affected. Interestingly, preformed vimentin filaments are more resistant to electrophile-induced disruption, although chemical modification is not diminished, showing that vimentin (lip)oxidation prior to assembly is more deleterious. In cells, electrophiles, particularly diamide, induce a fast and drastic disruption of existing filaments, which requires the presence of Cys328. As the cellular vimentin network is under continuous remodeling, we hypothesized that vimentin exchange on filaments would be necessary for diamide-induced disruption. We confirmed that strategies reducing vimentin dynamics, as monitored by FRAP, including cysteine crosslinking and ATP synthesis inhibition, prevent diamide effect. In turn, phosphorylation may promote vimentin disassembly. Indeed, treatment with the phosphatase inhibitor calyculin A to prevent dephosphorylation intensifies electrophile-induced wild-type vimentin filament disruption. However, whereas a phosphorylation-deficient vimentin mutant is only partially protected from disorganization, Cys328Ser vimentin is virtually resistant, even in the presence of calyculin A. Together, these results indicate that modification of Cys328 and vimentin exchange are critical for electrophile-induced network disruption.
Collapse
Affiliation(s)
- Andreia Mónico
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Sofia Duarte
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain; Molecular Hepatology Group, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
31
|
Furlani F, Sacco P, Scognamiglio F, Asaro F, Travan A, Borgogna M, Marsich E, Cok M, Paoletti S, Donati I. Nucleation, reorganization and disassembly of an active network from lactose-modified chitosan mimicking biological matrices. Carbohydr Polym 2019; 208:451-456. [DOI: 10.1016/j.carbpol.2018.12.096] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/20/2018] [Accepted: 12/30/2018] [Indexed: 01/07/2023]
|
32
|
Src and SHP2 coordinately regulate the dynamics and organization of vimentin filaments during cell migration. Oncogene 2019; 38:4075-4094. [PMID: 30696956 PMCID: PMC6755999 DOI: 10.1038/s41388-019-0705-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 11/28/2018] [Accepted: 01/04/2019] [Indexed: 12/27/2022]
Abstract
Vimentin intermediate filaments (VIFs), expressed in most mesenchymal and cancer cells, undergo dramatic reorganization during cell migration; however, the mechanism remains obscure. This study demonstrates that upon growth-factor stimulation, Src directly phosphorylates vimentin at Tyr117, leading to VIF disassembly into squiggles and particles at the cell edge during lamellipodia formation. The protein tyrosine phosphatase SHP2 counteracted the Src effects on VIF tyrosine phosphorylation and organization. VIFs formed by vimentin Y117D mutant were more soluble and dynamic than those formed by the wild-type and Y117F mutant. Increased expression of vimentin promoted growth-factor induced lamellipodia formation and cell migration, whereas the mutants suppressed both. The vimentin-induced increase in lamellipodia formation correlated with the activation of Rac and Vav2, with the latter associated with VIFs and recruited to the plasma membrane upon growth-factor stimulation. These results reveal a novel mechanism for regulating VIF dynamics through Src and SHP2 and demonstrate that proper VIF dynamics are important for Rac activation and cell migration.
Collapse
|
33
|
Abstract
The vimentin gene (
VIM) encodes one of the 71 human intermediate filament (IF) proteins, which are the building blocks of highly ordered, dynamic, and cell type-specific fiber networks. Vimentin is a multi-functional 466 amino acid protein with a high degree of evolutionary conservation among vertebrates.
Vim
−/− mice, though viable, exhibit systemic defects related to development and wound repair, which may have implications for understanding human disease pathogenesis. Vimentin IFs are required for the plasticity of mesenchymal cells under normal physiological conditions and for the migration of cancer cells that have undergone epithelial–mesenchymal transition. Although it was observed years ago that vimentin promotes cell migration, the molecular mechanisms were not completely understood. Recent advances in microscopic techniques, combined with computational image analysis, have helped illuminate vimentin dynamics and function in migrating cells on a precise scale. This review includes a brief historical account of early studies that unveiled vimentin as a unique component of the cell cytoskeleton followed by an overview of the physiological vimentin functions documented in studies on
Vim
−/− mice. The primary focus of the discussion is on novel mechanisms related to how vimentin coordinates cell migration. The current hypothesis is that vimentin promotes cell migration by integrating mechanical input from the environment and modulating the dynamics of microtubules and the actomyosin network. These new findings undoubtedly will open up multiple avenues to study the broader function of vimentin and other IF proteins in cell biology and will lead to critical insights into the relevance of different vimentin levels for the invasive behaviors of metastatic cancer cells.
Collapse
Affiliation(s)
- Rachel A Battaglia
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Samed Delic
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Harald Herrmann
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany.,Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Natasha T Snider
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
34
|
Favre B, Begré N, Bouameur JE, Lingasamy P, Conover GM, Fontao L, Borradori L. Desmoplakin interacts with the coil 1 of different types of intermediate filament proteins and displays high affinity for assembled intermediate filaments. PLoS One 2018; 13:e0205038. [PMID: 30286183 PMCID: PMC6171917 DOI: 10.1371/journal.pone.0205038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/18/2018] [Indexed: 12/04/2022] Open
Abstract
The interaction of intermediate filaments (IFs) with the cell-cell adhesion complexes desmosomes is crucial for cytoskeletal organization and cell resilience in the epidermis and heart. The intracellular desmosomal protein desmoplakin anchors IFs to the cell adhesion complexes predominantly via its four last carboxy-terminal domains (C-terminus). However, it remains unclear why the C-terminus of desmoplakin interacts with different IF types or if there are different binding affinities for each type of IFs that may influence the stability of cell-specific adhesion complexes. By yeast three-hybrid and fluorescence binding assays, we found that the coiled-coil 1 of the conserved central rod domain of the heterodimeric cytokeratins (Ks) 5 and 14 (K5/K14) was required for their interaction with the C-terminus of desmoplakin, while their unique amino head- and C-tail domains were dispensable. Similar findings were obtained in vitro with K1/K10, and the type III IF proteins desmin and vimentin. Binding assays testing the C-terminus of desmoplakin with assembled K5/K14 and desmin IFs yielded an apparent affinity in the nM range. Our findings reveal that the same conserved domain of IF proteins binds to the C-terminus of desmoplakin, which may help explain the previously reported broad binding IF-specificity to desmoplakin. Our data suggest that desmoplakin high-affinity binding to diverse IF proteins ensures robust linkages of IF cytoskeleton and desmosomes that maintain the structural integrity of cellular adhesion complexes. In summary, our results give new insights into the molecular basis of the IF-desmosome association.
Collapse
Affiliation(s)
- Bertrand Favre
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Nadja Begré
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Jamal-Eddine Bouameur
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Prakash Lingasamy
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for Biomedical Research, University of Bern, Bern, Switzerland
| | - Gloria M. Conover
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - Lionel Fontao
- Department of Dermatology, Geneva University Hospitals, Geneva, Switzerland
| | - Luca Borradori
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| |
Collapse
|
35
|
Danielsson F, Peterson MK, Caldeira Araújo H, Lautenschläger F, Gad AKB. Vimentin Diversity in Health and Disease. Cells 2018; 7:E147. [PMID: 30248895 PMCID: PMC6210396 DOI: 10.3390/cells7100147] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022] Open
Abstract
Vimentin is a protein that has been linked to a large variety of pathophysiological conditions, including cataracts, Crohn's disease, rheumatoid arthritis, HIV and cancer. Vimentin has also been shown to regulate a wide spectrum of basic cellular functions. In cells, vimentin assembles into a network of filaments that spans the cytoplasm. It can also be found in smaller, non-filamentous forms that can localise both within cells and within the extracellular microenvironment. The vimentin structure can be altered by subunit exchange, cleavage into different sizes, re-annealing, post-translational modifications and interacting proteins. Together with the observation that different domains of vimentin might have evolved under different selection pressures that defined distinct biological functions for different parts of the protein, the many diverse variants of vimentin might be the cause of its functional diversity. A number of review articles have focussed on the biology and medical aspects of intermediate filament proteins without particular commitment to vimentin, and other reviews have focussed on intermediate filaments in an in vitro context. In contrast, the present review focusses almost exclusively on vimentin, and covers both ex vivo and in vivo data from tissue culture and from living organisms, including a summary of the many phenotypes of vimentin knockout animals. Our aim is to provide a comprehensive overview of the current understanding of the many diverse aspects of vimentin, from biochemical, mechanical, cellular, systems biology and medical perspectives.
Collapse
Affiliation(s)
- Frida Danielsson
- Science for Life Laboratory, Royal Institute of Technology, 17165 Stockholm, Sweden.
| | | | | | - Franziska Lautenschläger
- Campus D2 2, Leibniz-Institut für Neue Materialien gGmbH (INM) and Experimental Physics, NT Faculty, E 2 6, Saarland University, 66123 Saarbrücken, Germany.
| | - Annica Karin Britt Gad
- Centro de Química da Madeira, Universidade da Madeira, 9020105 Funchal, Portugal.
- Department of Medical Biochemistry and Microbiology, Uppsala University, 75237 Uppsala, Sweden.
| |
Collapse
|
36
|
Robert A, Tian P, Adam SA, Kittisopikul M, Jaqaman K, Goldman RD, Gelfand VI. Kinesin-dependent transport of keratin filaments: a unified mechanism for intermediate filament transport. FASEB J 2018; 33:388-399. [PMID: 29944446 PMCID: PMC6355078 DOI: 10.1096/fj.201800604r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Keratin intermediate filaments (IFs) are the major cytoskeletal component in epithelial cells. The dynamics of keratin IFs have been described to depend mostly on the actin cytoskeleton, but the rapid transport of fully polymerized keratin filaments has not been reported. In this work, we used a combination of photoconversion experiments and clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeats-associated protein 9 genome editing to study the role of microtubules and microtubule motors in keratin filament transport. We found that long keratin filaments, like other types of IFs, are transported along microtubules by kinesin-1. Our data revealed that keratin and vimentin are nonconventional kinesin-1 cargoes because their transport did not require kinesin light chains, which are a typical adapter for kinesin-dependent cargo transport. Furthermore, we found that the same domain of the kinesin heavy chain tail is involved in keratin and vimentin IF transport, strongly suggesting that multiple types of IFs move along microtubules using an identical mechanism.-Robert, A., Tian, P., Adam, S. A., Kittisopikul, M., Jaqaman, K., Goldman, R. D., Gelfand, V. I. Kinesin-dependent transport of keratin filaments: a unified mechanism for intermediate filament transport.
Collapse
Affiliation(s)
- Amélie Robert
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Peirun Tian
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Stephen A Adam
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mark Kittisopikul
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA; and
| | - Khuloud Jaqaman
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, Texas, USA; and.,Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Robert D Goldman
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
37
|
Lee YA, Kim JJ, Lee J, Lee JHJ, Sahu S, Kwon HY, Park SJ, Jang SY, Lee JS, Wang Z, Tam WL, Lim B, Kang NY, Chang YT. Identification of Tumor Initiating Cells with a Small-Molecule Fluorescent Probe by Using Vimentin as a Biomarker. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201712920] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Yong-An Lee
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
| | - Jong-Jin Kim
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
| | - Jungyeol Lee
- Department of Chemistry; Pohang University of Science and Technology; Pohang 37673 Korea
- Present address: New drug discovery center; DGMIF; Daegu 41061 Korea
| | - Jia Hui Jane Lee
- Genome Institute of Singapore; Agency for Science Technology and Research (A*STAR); Singapore 138672 Singapore
- School of Biological Sciences; Nanyang Technological University; Singapore 637551 Singapore
| | - Srikanta Sahu
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
| | - Haw-Young Kwon
- Center for Self-assembly and Complexity; Institute for Basic Science (IBS); Pohang 37673 Korea
| | - Sung-Jin Park
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
| | - Se-Young Jang
- Molecular Recognition Research Center; Korea Institute of Science and Technology; Seoul 02792 Korea
| | - Jun-Seok Lee
- Molecular Recognition Research Center; Korea Institute of Science and Technology; Seoul 02792 Korea
| | - Zhenxun Wang
- Genome Institute of Singapore; Agency for Science Technology and Research (A*STAR); Singapore 138672 Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore; Agency for Science Technology and Research (A*STAR); Singapore 138672 Singapore
- Cancer Science Institute of Singapore; National University of Singapore; Singapore 117599 Singapore
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore 117596 Singapore
| | - Bing Lim
- Genome Institute of Singapore; Agency for Science Technology and Research (A*STAR); Singapore 138672 Singapore
- Present address: Merck Sharp and Dohme Translational Medicine Research Center; Singapore 138648 Singapore
| | - Nam-Young Kang
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
- Present address: New drug discovery center; DGMIF; Daegu 41061 Korea
| | - Young-Tae Chang
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
- Department of Chemistry; Pohang University of Science and Technology; Pohang 37673 Korea
- Center for Self-assembly and Complexity; Institute for Basic Science (IBS); Pohang 37673 Korea
| |
Collapse
|
38
|
Lee YA, Kim JJ, Lee J, Lee JHJ, Sahu S, Kwon HY, Park SJ, Jang SY, Lee JS, Wang Z, Tam WL, Lim B, Kang NY, Chang YT. Identification of Tumor Initiating Cells with a Small-Molecule Fluorescent Probe by Using Vimentin as a Biomarker. Angew Chem Int Ed Engl 2018; 57:2851-2854. [DOI: 10.1002/anie.201712920] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/16/2018] [Indexed: 01/06/2023]
Affiliation(s)
- Yong-An Lee
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
| | - Jong-Jin Kim
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
| | - Jungyeol Lee
- Department of Chemistry; Pohang University of Science and Technology; Pohang 37673 Korea
- Present address: New drug discovery center; DGMIF; Daegu 41061 Korea
| | - Jia Hui Jane Lee
- Genome Institute of Singapore; Agency for Science Technology and Research (A*STAR); Singapore 138672 Singapore
- School of Biological Sciences; Nanyang Technological University; Singapore 637551 Singapore
| | - Srikanta Sahu
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
| | - Haw-Young Kwon
- Center for Self-assembly and Complexity; Institute for Basic Science (IBS); Pohang 37673 Korea
| | - Sung-Jin Park
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
| | - Se-Young Jang
- Molecular Recognition Research Center; Korea Institute of Science and Technology; Seoul 02792 Korea
| | - Jun-Seok Lee
- Molecular Recognition Research Center; Korea Institute of Science and Technology; Seoul 02792 Korea
| | - Zhenxun Wang
- Genome Institute of Singapore; Agency for Science Technology and Research (A*STAR); Singapore 138672 Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore; Agency for Science Technology and Research (A*STAR); Singapore 138672 Singapore
- Cancer Science Institute of Singapore; National University of Singapore; Singapore 117599 Singapore
- Department of Biochemistry; Yong Loo Lin School of Medicine; National University of Singapore; Singapore 117596 Singapore
| | - Bing Lim
- Genome Institute of Singapore; Agency for Science Technology and Research (A*STAR); Singapore 138672 Singapore
- Present address: Merck Sharp and Dohme Translational Medicine Research Center; Singapore 138648 Singapore
| | - Nam-Young Kang
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
- Present address: New drug discovery center; DGMIF; Daegu 41061 Korea
| | - Young-Tae Chang
- Singapore Bioimaging Consortium; Agency for Science Technology and Research (A*STAR); Singapore 138667 Singapore
- Department of Chemistry; Pohang University of Science and Technology; Pohang 37673 Korea
- Center for Self-assembly and Complexity; Institute for Basic Science (IBS); Pohang 37673 Korea
| |
Collapse
|
39
|
Leube RE, Moch M, Windoffer R. Intracellular Motility of Intermediate Filaments. Cold Spring Harb Perspect Biol 2017; 9:9/6/a021980. [PMID: 28572456 DOI: 10.1101/cshperspect.a021980] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
SUMMARYThe establishment and continuous cell type-specific adaptation of cytoplasmic intermediate filament (IF) networks are linked to various types of IF motility. Motor protein-driven active transport, linkage to other cellular structures, diffusion of small soluble subunits, and intrinsic network elasticity all contribute to the motile behavior of IFs. These processes are subject to regulation by multiple signaling pathways. IF motility is thereby connected to and involved in many basic cellular processes guarding the maintenance of cell and tissue integrity. Disturbances of IF motility are linked to diseases that are characterized by cytoplasmic aggregates containing IF proteins together with other cellular components.
Collapse
Affiliation(s)
- Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Marcin Moch
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Reinhard Windoffer
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
40
|
Microtubule-microtubule sliding by kinesin-1 is essential for normal cytoplasmic streaming in Drosophila oocytes. Proc Natl Acad Sci U S A 2016; 113:E4995-5004. [PMID: 27512034 DOI: 10.1073/pnas.1522424113] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cytoplasmic streaming in Drosophila oocytes is a microtubule-based bulk cytoplasmic movement. Streaming efficiently circulates and localizes mRNAs and proteins deposited by the nurse cells across the oocyte. This movement is driven by kinesin-1, a major microtubule motor. Recently, we have shown that kinesin-1 heavy chain (KHC) can transport one microtubule on another microtubule, thus driving microtubule-microtubule sliding in multiple cell types. To study the role of microtubule sliding in oocyte cytoplasmic streaming, we used a Khc mutant that is deficient in microtubule sliding but able to transport a majority of cargoes. We demonstrated that streaming is reduced by genomic replacement of wild-type Khc with this sliding-deficient mutant. Streaming can be fully rescued by wild-type KHC and partially rescued by a chimeric motor that cannot move organelles but is active in microtubule sliding. Consistent with these data, we identified two populations of microtubules in fast-streaming oocytes: a network of stable microtubules anchored to the actin cortex and free cytoplasmic microtubules that moved in the ooplasm. We further demonstrated that the reduced streaming in sliding-deficient oocytes resulted in posterior determination defects. Together, we propose that kinesin-1 slides free cytoplasmic microtubules against cortically immobilized microtubules, generating forces that contribute to cytoplasmic streaming and are essential for the refinement of posterior determinants.
Collapse
|
41
|
Robert A, Hookway C, Gelfand VI. Intermediate filament dynamics: What we can see now and why it matters. Bioessays 2016; 38:232-43. [PMID: 26763143 DOI: 10.1002/bies.201500142] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The mechanical properties of vertebrate cells are largely defined by the system of intermediate filaments (IF). As part of a dense network, IF polymers are constantly rearranged and relocalized in the cell to fulfill their duty as cells change shape, migrate, or divide. With the development of new imaging technologies, such as photoconvertible proteins and super-resolution microscopy, a new appreciation for the complexity of IF dynamics has emerged. This review highlights new findings about the transport of IF, the remodeling of filaments by a process of severing and re-annealing, and the subunit exchange that occurs between filament precursors and a soluble pool of IF. We will also discuss the unique dynamic features of the keratin IF network. Finally, we will speculate about how the dynamic properties of IF are related to their functions.
Collapse
Affiliation(s)
- Amélie Robert
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Caroline Hookway
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
42
|
Ridge KM, Shumaker D, Robert A, Hookway C, Gelfand VI, Janmey PA, Lowery J, Guo M, Weitz DA, Kuczmarski E, Goldman RD. Methods for Determining the Cellular Functions of Vimentin Intermediate Filaments. Methods Enzymol 2015; 568:389-426. [PMID: 26795478 DOI: 10.1016/bs.mie.2015.09.036] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The type III intermediate filament protein vimentin was once thought to function mainly as a static structural protein in the cytoskeleton of cells of mesenchymal origin. Now, however, vimentin is known to form a dynamic, flexible network that plays an important role in a number of signaling pathways. Here, we describe various methods that have been developed to investigate the cellular functions of the vimentin protein and intermediate filament network, including chemical disruption, photoactivation and photoconversion, biolayer interferometry, soluble bead binding assay, three-dimensional substrate experiments, collagen gel contraction, optical-tweezer active microrheology, and force spectrum microscopy. Using these techniques, the contributions of vimentin to essential cellular processes can be probed in ever further detail.
Collapse
Affiliation(s)
- Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Chicago, Illinois, USA; Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA; Veterans Administration, Chicago, Illinois, USA.
| | - Dale Shumaker
- Division of Pulmonary and Critical Care Medicine, Chicago, Illinois, USA; Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Amélie Robert
- Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Caroline Hookway
- Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Vladimir I Gelfand
- Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Paul A Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Departments of Physiology and Physics & Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jason Lowery
- Division of Pulmonary and Critical Care Medicine, Chicago, Illinois, USA
| | - Ming Guo
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - David A Weitz
- School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA; Department of Physics, Harvard University, Cambridge, Massachusetts, USA
| | - Edward Kuczmarski
- Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Robert D Goldman
- Division of Pulmonary and Critical Care Medicine, Chicago, Illinois, USA; Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
43
|
Abstract
Withaferin A (WFA), initially identified as a compound that inhibits experimental angiogenesis, has been shown to bind to soluble vimentin (sVim) and other type III intermediate filament (IF) proteins. We review WFA's dose-related activities (Section 1), examining nanomolar concentrations effects on sVim in cell proliferation and submicromolar effects on lamellipodia and focal adhesion formation. WFA effects on polymeric IFs are especially interesting to the study of cell migration and invasion that depend on IF mechanical contractile properties. WFA interferes with NF-κB signaling, though this anti-inflammatory mechanism may occur via perturbation of sVim-protein complexes, and possibly also via targeting IκB kinase β directly. However, micromolar concentrations that induce vimentin cleavage to promote apoptosis may increasingly show off-target effects via targeting other IFs (neurofilaments and keratin) and non-IFs (tubulin, heat-shock proteins, proteasome). Thus, in Section 2, we describe our studies combining cell cultures with animal models of injury to validate relevant type III IF-targeting mechanisms of WFA. In Section 3, we illuminate from investigating myofibroblast differentiation how sVim phosphorylation may govern cell type-selective sensitivity to WFA, offering impetus for exploring vimentin phosphorylation isoforms as targets and biomarkers of fibrosis. These different WFA targets and activities are listed in a summary table.
Collapse
Affiliation(s)
- Royce Mohan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, USA.
| | - Paola Bargagna-Mohan
- Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|