1
|
Renganathan B, Moore AS, Yeo WH, Petruncio A, Ackerman D, Weigel AV, Team TC, Pasolli HA, Xu CS, Shtengel G, Hess HF, Serpinskaya AS, Zhang HF, Lippincott-Schwartz J, Gelfand VI. Vimentin filament transport and organization revealed by single-particle tracking and 3D FIB-SEM. J Cell Biol 2025; 224:e202406054. [PMID: 40062969 PMCID: PMC11893169 DOI: 10.1083/jcb.202406054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/22/2024] [Accepted: 01/09/2025] [Indexed: 03/14/2025] Open
Abstract
Vimentin intermediate filaments (VIFs) form complex, tightly packed networks; due to this density, traditional imaging approaches cannot discern single-filament behavior. To address this, we developed and validated a sparse vimentin-SunTag labeling strategy, enabling single-particle tracking of individual VIFs and providing a sensitive, unbiased, and quantitative method for measuring global VIF motility. Using this approach, we define the steady-state VIF motility rate, showing a constant ∼8% of VIFs undergo directed microtubule-based motion irrespective of subcellular location or local filament density. Significantly, our single-particle tracking approach revealed uncorrelated motion of individual VIFs within bundles, an observation seemingly at odds with conventional models of tightly cross-linked bundles. To address this, we acquired high-resolution focused ion beam scanning electron microscopy volumes of vitreously frozen cells and reconstructed three-dimensional VIF bundles, finding that they form only loosely organized, semi-coherent structures from which single VIFs frequently emerge to locally engage neighboring microtubules. Overall, this work demonstrates single VIF dynamics and organization in the cellular milieu for the first time.
Collapse
Affiliation(s)
- Bhuvanasundar Renganathan
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Andrew S. Moore
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Wei-Hong Yeo
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Alyson Petruncio
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - David Ackerman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Aubrey V. Weigel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - The CellMap Team
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - H. Amalia Pasolli
- Electron Microscopy Resource Center, Rockefeller University, New York, NY, USA
| | - C. Shan Xu
- Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Gleb Shtengel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Harald F. Hess
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Anna S. Serpinskaya
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hao F. Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | | | - Vladimir I. Gelfand
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
2
|
Rosier M, Krstulović A, Kim HR, Kaur N, Enakireru EM, Symmes D, Dobra K, Chen R, Evans CA, Gad AKB. The Vimentin-Targeting Drug ALD-R491 Partially Reverts the Epithelial-to-Mesenchymal Transition and Vimentin Interactome of Lung Cancer Cells. Cancers (Basel) 2024; 17:81. [PMID: 39796712 PMCID: PMC11720119 DOI: 10.3390/cancers17010081] [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: 11/17/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Background: The epithelial-to-mesenchymal transition (EMT) is a common feature in early cancer invasion. Increased vimentin is a canonical marker of the EMT; however, the role of vimentin in EMT remains unknown. Methods: To clarify this, we induced EMT in lung cancer cells with TGF-β1, followed by treatment with the vimentin-targeting drug ALD-R491, live-cell imaging, and quantitative proteomics. Results: We identified 838 proteins in the intermediate filament fraction of cells. TGF-β1 treatment increased the proportion of vimentin in this fraction and the levels of 24 proteins. Variants of fibronectin showed the most pronounced increase (137-fold), followed by regulators of the cytoskeleton, cell motility, and division, such as the mRNA-splicing protein SON. TGF-β1 increased cell spreading and cell migration speed, and changed a positive correlation between cell migration speed and persistence to negative. ALD-R491 reversed these mesenchymal phenotypes to epithelial and the binding of RNA-binding proteins, including SON. Conclusions: These findings present many new interactors of intermediate filaments, describe how EMT and vimentin filament dynamics influence the intermediate filament interactome, and present ALD-R491 as a possible EMT-inhibitor. The observations support the hypothesis that the dynamic turnover of vimentin filaments and their interacting proteins govern mesenchymal cell migration, EMT, cell invasion, and cancer metastasis.
Collapse
Affiliation(s)
- Marieke Rosier
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden; (M.R.); (A.K.); (N.K.); (E.M.E.); (K.D.)
| | - Anja Krstulović
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden; (M.R.); (A.K.); (N.K.); (E.M.E.); (K.D.)
| | - Hyejeong Rosemary Kim
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield S10 2RX, UK;
| | - Nihardeep Kaur
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden; (M.R.); (A.K.); (N.K.); (E.M.E.); (K.D.)
| | - Erhumuoghene Mary Enakireru
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden; (M.R.); (A.K.); (N.K.); (E.M.E.); (K.D.)
| | - Deebie Symmes
- Aluda Pharmaceuticals, Inc., Menlo Park, CA 94025, USA; (D.S.); (R.C.)
| | - Katalin Dobra
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden; (M.R.); (A.K.); (N.K.); (E.M.E.); (K.D.)
| | - Ruihuan Chen
- Aluda Pharmaceuticals, Inc., Menlo Park, CA 94025, USA; (D.S.); (R.C.)
| | - Caroline A. Evans
- School of Materials, Chemical and Biological Engineering, University of Sheffield, Sheffield S10 2TN, UK;
| | - Annica K. B. Gad
- Department of Oncology-Pathology, Karolinska Institutet, 171 64 Solna, Sweden; (M.R.); (A.K.); (N.K.); (E.M.E.); (K.D.)
| |
Collapse
|
3
|
Hu X, Jalal S, Yao M, Bakke O, Margadant F, Sheetz M. Differential Talin cleavage in transformed and non-transformed cells and its consequences. Front Cell Dev Biol 2024; 12:1430728. [PMID: 39086658 PMCID: PMC11289324 DOI: 10.3389/fcell.2024.1430728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
This study investigates differences in focal adhesion (FA) morphology and Talin cleavage levels between transformed and non-transformed cell lines. Utilizing fluorescently tagged wild-type Talin and Talin mutants with calpain cleavage site mutations, FA structures were visualized. Mutations in different Talin cleavage sites showed varying impacts on FA morphology and distribution across melanoma cell lines (Meljuso, A375P, A2058) and a non-transformed cell line (HFF). Western blot analysis, ratiometric fluorescence intensity-based measurements, and FRAP experiments revealed higher Talin cleavage levels within FAs of transformed cell lines compared to non-transformed cells. Additionally, growth assays indicated that reducing calpain cleavage levels attenuated transformed cell growth. These findings suggest that Talin cleavage level is crucial for FA morphology and assembly, with higher levels observed in transformed cells, influencing their growth dynamics.
Collapse
Affiliation(s)
- Xian Hu
- Center for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Salma Jalal
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
| | - Mingxi Yao
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Oddmund Bakke
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Felix Margadant
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX, United States
| | - Michael Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Molecular Mechanomedicine Program, Biochemistry and Molecular Biology Department, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
4
|
Renganathan B, Moore A, Yeo WH, Petruncio A, Ackerman D, Wiegel A, The CellMap Team, Pasolli HA, Xu CS, Shtengel G, Hess HF, Serpinskaya AS, Zhang HF, Lippincott-Schwartz J, Gelfand VI. Transport and Organization of Individual Vimentin Filaments Within Dense Networks Revealed by Single Particle Tracking and 3D FIB-SEM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598346. [PMID: 38915582 PMCID: PMC11195130 DOI: 10.1101/2024.06.10.598346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Single-particle tracking demonstrates that individual filaments in bundles of vimentin intermediate filaments are transported in the cytoplasm by motor proteins along microtubules. Furthermore, using 3D FIB-SEM the authors showed that vimentin filament bundles are loosely packed and coaligned with microtubules. Vimentin intermediate filaments (VIFs) form complex, tight-packed networks; due to this density, traditional ensemble labeling and imaging approaches cannot accurately discern single filament behavior. To address this, we introduce a sparse vimentin-SunTag labeling strategy to unambiguously visualize individual filament dynamics. This technique confirmed known long-range dynein and kinesin transport of peripheral VIFs and uncovered extensive bidirectional VIF motion within the perinuclear vimentin network, a region we had thought too densely bundled to permit such motility. To examine the nanoscale organization of perinuclear vimentin, we acquired high-resolution electron microscopy volumes of a vitreously frozen cell and reconstructed VIFs and microtubules within a ~50 μm3 window. Of 583 VIFs identified, most were integrated into long, semi-coherent bundles that fluctuated in width and filament packing density. Unexpectedly, VIFs displayed minimal local co-alignment with microtubules, save for sporadic cross-over sites that we predict facilitate cytoskeletal crosstalk. Overall, this work demonstrates single VIF dynamics and organization in the cellular milieu for the first time.
Collapse
Affiliation(s)
- Bhuvanasundar Renganathan
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Andrew Moore
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Wei-Hong Yeo
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60201, USA
| | - Alyson Petruncio
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - David Ackerman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Aubrey Wiegel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - The CellMap Team
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - H. Amalia Pasolli
- Electron Microscopy Resource Center, Rockefeller University, New York, NY 10021, USA
| | - C. Shan Xu
- Department of Cellular and Molecular Physiology, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Gleb Shtengel
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Harald F. Hess
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Anna S Serpinskaya
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Hao F. Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60201, USA
| | | | - Vladimir I. Gelfand
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
5
|
Hakibilen C, Delort F, Daher MT, Joanne P, Cabet E, Cardoso O, Bourgois-Rocha F, Tian C, Rivas E, Madruga M, Ferreiro A, Lilienbaum A, Vicart P, Agbulut O, Hénon S, Batonnet-Pichon S. Desmin Modulates Muscle Cell Adhesion and Migration. Front Cell Dev Biol 2022; 10:783724. [PMID: 35350386 PMCID: PMC8957967 DOI: 10.3389/fcell.2022.783724] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 02/09/2022] [Indexed: 12/04/2022] Open
Abstract
Cellular adhesion and migration are key functions that are disrupted in numerous diseases. We report that desmin, a type-III muscle-specific intermediate filament, is a novel cell adhesion regulator. Expression of p.R406W mutant desmin, identified in patients with desmin-related myopathy, modified focal adhesion area and expression of adhesion-signaling genes in myogenic C2C12 cells. Satellite cells extracted from desmin-knock-out (DesKO) and desmin-knock-in-p.R405W (DesKI-R405W) mice were less adhesive and migrated faster than those from wild-type mice. Moreover, we observed mislocalized and aggregated vinculin, a key component of cell adhesion, in DesKO and DesKI-R405W muscles. Vinculin expression was also increased in desmin-related myopathy patient muscles. Together, our results establish a novel role for desmin in cell-matrix adhesion, an essential process for strength transmission, satellite cell migration and muscle regeneration. Our study links the patho-physiological mechanisms of desminopathies to adhesion/migration defects, and may lead to new cellular targets for novel therapeutic approaches.
Collapse
Affiliation(s)
| | | | | | - Pierre Joanne
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Eva Cabet
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France
| | | | | | - Cuixia Tian
- Department of Neurology, Cincinnati Children's Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Eloy Rivas
- Servicio de Anatomia Patologica, Hospital Universitario Virgen Del Rocio, Sevilla, Spain
| | - Marcos Madruga
- Unidad de Neurologia Pediatrica, Hospital Universitario Virgen Del Rocio, Sevilla, Spain
| | - Ana Ferreiro
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France.,APHP, Centre de Référence Maladies Neuromusculaires Nord/Est/Ile-de-France, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | | | - Patrick Vicart
- Université de Paris, BFA, UMR 8251, CNRS, Paris, France.,Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France.,Université de Paris, MSC, UMR 7067, CNRS, Paris, France.,Department of Neurology, Cincinnati Children's Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, United States.,Servicio de Anatomia Patologica, Hospital Universitario Virgen Del Rocio, Sevilla, Spain.,Unidad de Neurologia Pediatrica, Hospital Universitario Virgen Del Rocio, Sevilla, Spain.,APHP, Centre de Référence Maladies Neuromusculaires Nord/Est/Ile-de-France, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Onnik Agbulut
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), CNRS UMR 8256, INSERM ERL U1164, Biological Adaptation and Ageing, Paris, France
| | - Sylvie Hénon
- Université de Paris, MSC, UMR 7067, CNRS, Paris, France
| | | |
Collapse
|
6
|
Evans CA, Kim HR, Macfarlane SC, Nowicki PIA, Baltes C, Xu L, Widengren J, Lautenschläger F, Corfe BM, Gad AKB. Metastasising Fibroblasts Show an HDAC6-Dependent Increase in Migration Speed and Loss of Directionality Linked to Major Changes in the Vimentin Interactome. Int J Mol Sci 2022; 23:1961. [PMID: 35216078 PMCID: PMC8880509 DOI: 10.3390/ijms23041961] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/07/2023] Open
Abstract
Metastasising cells express the intermediate filament protein vimentin, which is used to diagnose invasive tumours in the clinic. We aimed to clarify how vimentin regulates the motility of metastasising fibroblasts. STED super-resolution microscopy, live-cell imaging and quantitative proteomics revealed that oncogene-expressing and metastasising fibroblasts show a less-elongated cell shape, reduced cell spreading, increased cell migration speed, reduced directionality, and stronger coupling between these migration parameters compared to normal control cells. In total, we identified and compared 555 proteins in the vimentin interactome. In metastasising cells, the levels of keratin 18 and Rab5C were increased, while those of actin and collagen were decreased. Inhibition of HDAC6 reversed the shape, spreading and migration phenotypes of metastasising cells back to normal. Inhibition of HDAC6 also decreased the levels of talin 1, tropomyosin, Rab GDI β, collagen and emilin 1 in the vimentin interactome, and partially reversed the nanoscale vimentin organisation in oncogene-expressing cells. These findings describe the changes in the vimentin interactome and nanoscale distribution that accompany the defective cell shape, spreading and migration of metastasising cells. These results support the hypothesis that oncogenes can act through HDAC6 to regulate the vimentin binding of the cytoskeletal and cell-extracellular matrix adhesion components that contribute to the defective motility of metastasising cells.
Collapse
Affiliation(s)
- Caroline A. Evans
- Department of Chemical and Biological Engineering, University of Sheffield, Mappin St, Sheffield S1 3JD, UK;
| | - Hyejeong Rosemary Kim
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (H.R.K.); (S.C.M.); (P.I.A.N.)
| | - Sarah C. Macfarlane
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (H.R.K.); (S.C.M.); (P.I.A.N.)
| | - Poppy I. A. Nowicki
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (H.R.K.); (S.C.M.); (P.I.A.N.)
| | - Carsten Baltes
- Experimental Physics, NT Faculty, D2 2, Saarland University, 66123 Saarbrücken, Germany; (C.B.); (F.L.)
| | - Lei Xu
- Department of Applied Physics/Experimental Biomolecular Physics, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden; (L.X.); (J.W.)
| | - Jerker Widengren
- Department of Applied Physics/Experimental Biomolecular Physics, KTH Royal Institute of Technology, SE-100 44 Stockholm, Sweden; (L.X.); (J.W.)
| | - Franziska Lautenschläger
- Experimental Physics, NT Faculty, D2 2, Saarland University, 66123 Saarbrücken, Germany; (C.B.); (F.L.)
| | - Bernard M. Corfe
- Population Health Sciences Institute, Human Nutrition Research Centre, Faculty of Medical Sciences, Newcastle University, Newcastle NE2 4HH, UK;
| | - Annica K. B. Gad
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (H.R.K.); (S.C.M.); (P.I.A.N.)
- Madeira Chemistry Research Centre, University of Madeira, 9020105 Funchal, Portugal
| |
Collapse
|
7
|
Liang Y, Li L, Chen Y, Zhang S, Li Z, Xiao J, Wei D. Research Progress on the Role of Intermediate Filament Vimentin in Atherosclerosis. DNA Cell Biol 2021; 40:1495-1502. [PMID: 34931866 DOI: 10.1089/dna.2021.0623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The cytoskeleton is a biopolymer network composed of intermediate filaments, actin, and microtubules, which is the main mechanical structure of cells. Vimentin is an intermediate filament protein that regulates the mechanical and contractile properties of cells, thereby reflecting their mechanical properties. In recent years, the "nonmechanical function" of vimentin inside and outside of cells has attracted extensive attention. The content of vimentin in atherosclerotic plaques is increased, and the serum secretion of vimentin in patients with coronary heart disease is remarkably increased. In this review, the mechanistic and nonmechanistic roles of vimentin in atherosclerosis progression were summarized on the basis of current studies.
Collapse
Affiliation(s)
- Yamin Liang
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Lu Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Yanmei Chen
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Shulei Zhang
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhaozhi Li
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Jinyan Xiao
- YueYang Maternal-Child Medicine Health Hospital Hunan Province Innovative Training Base for Medical Postgraduates, University of China South China and Yueyang Women and Children's Medical Center, Yueyang, Hunan, China
| | - Dangheng Wei
- Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Institute of Cardiovascular Disease, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| |
Collapse
|
8
|
Xie YH, Tang CQ, Huang ZZ, Zhou SC, Yang YW, Yin Z, Heng BC, Chen WS, Chen X, Shen WL. ECM remodeling in stem cell culture: a potential target for regulating stem cell function. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:542-554. [PMID: 34082581 DOI: 10.1089/ten.teb.2021.0066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Stem cells (SCs) hold great potential for regenerative medicine, tissue engineering and cell therapy. The clinical applications of SCs require both high quality and quantity of transplantable cells. However, during conventional in vitro expansion, SCs tend to lose properties that make them amenable for cell therapies. Extracellular matrix (ECM) serves an essential regulatory part in the growth, differentiation and homeostasis of all cells in vivo. when signals transmitted to cells, they do not respond passively. Many cell types can remodel pericellular matrix to meet their specific needs. This reciprocal cell-ECM interaction is crucial for the conservation of cell and tissue functions and homeostasis. In vitro ECM remodeling also plays a key role in regulating the lineage fate of SCs. A deeper understanding of in vitro ECM remodeling may provide new perspectives for the maintenance of SC function. In this review, we critically examined three ways that cells can be used to influence the pericellular matrix: (i) exerting tensile force on the ECM, (ii) secreting a variety of ECM proteins, and (iii) degrading the surrounding matrix, and its impact on SC lineage fate. Finally, we describe the deficiencies of current studies and what needs to be done next to further understand the role of ECM remodeling in ex vivo SC cultures.
Collapse
Affiliation(s)
- Yuan-Hao Xie
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| | - Chen-Qi Tang
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| | - Zi-Zhan Huang
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| | - Si-Cheng Zhou
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, Zhejiang, China;
| | - Yu-Wei Yang
- Zhejiang University School of Medicine, 26441, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Hangzhou, Zhejiang, China;
| | - Zi Yin
- Zhejiang University School of Medicine, 26441, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Hangzhou, Zhejiang, China;
| | - Boon Chin Heng
- Peking University School of Stomatology, 159460, Beijing, Beijing, China;
| | - Wei-Shan Chen
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| | - Xiao Chen
- Zhejiang University School of Medicine, 26441, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Hangzhou, Zhejiang, China;
| | - Wei-Liang Shen
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| |
Collapse
|
9
|
Del Favero G, Zeugswetter M, Kiss E, Marko D. Endoplasmic Reticulum Adaptation and Autophagic Competence Shape Response to Fluid Shear Stress in T24 Bladder Cancer Cells. Front Pharmacol 2021; 12:647350. [PMID: 34012396 PMCID: PMC8126838 DOI: 10.3389/fphar.2021.647350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 12/26/2022] Open
Abstract
Accumulation of xenobiotics and waste metabolites in the urinary bladder is constantly accompanied by shear stress originating from the movement of the luminal fluids. Hence, both chemical and physical cues constantly modulate the cellular response in health and disease. In line, bladder cells have to maintain elevated mechanosensory competence together with chemical stress response adaptation potential. However, much of the molecular mechanisms sustaining this plasticity is currently unknown. Taking this as a starting point, we investigated the response of T24 urinary bladder cancer cells to shear stress comparing morphology to functional performance. T24 cells responded to the shear stress protocol (flow speed of 0.03 ml/min, 3 h) by significantly increasing their surface area. When exposed to deoxynivalenol-3-sulfate (DON-3-Sulf), bladder cells increased this response in a concentration-dependent manner (0.1-1 µM). DON-3-Sulf is a urinary metabolite of a very common food contaminant mycotoxin (deoxynivalenol, DON) and was already described to enhance proliferation of cancer cells. Incubation with DON-3-Sulf also caused the enlargement of the endoplasmic reticulum (ER), decreased the lysosomal movement, and increased the formation of actin stress fibers. Similar remodeling of the endoplasmic reticulum and area spread after shear stress were observed upon incubation with the autophagy activator rapamycin (1-100 nM). Performance of experiments in the presence of chloroquine (chloroquine, 30 μM) further contributed to shed light on the mechanistic link between adaptation to the biomechanical stimulation and ER stress response. At the molecular level, we observed that ER reshaping was linked to actin organization, with the two components mutually regulating each other. Indeed, we identified in the ER stress-cytoskeletal rearrangement an important axis defining the physical/chemical response potential of bladder cells and created a workflow for further investigation of urinary metabolites, food constituents, and contaminants, as well as for pharmacological profiling.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria.,Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Michael Zeugswetter
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Sjöqvist M, Antfolk D, Suarez-Rodriguez F, Sahlgren C. From structural resilience to cell specification - Intermediate filaments as regulators of cell fate. FASEB J 2020; 35:e21182. [PMID: 33205514 PMCID: PMC7839487 DOI: 10.1096/fj.202001627r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 12/18/2022]
Abstract
During the last decades intermediate filaments (IFs) have emerged as important regulators of cellular signaling events, ascribing IFs with functions beyond the structural support they provide. The organ and developmental stage‐specific expression of IFs regulate cell differentiation within developing or remodeling tissues. Lack of IFs causes perturbed stem cell differentiation in vasculature, intestine, nervous system, and mammary gland, in transgenic mouse models. The aberrant cell fate decisions are caused by deregulation of different stem cell signaling pathways, such as Notch, Wnt, YAP/TAZ, and TGFβ. Mutations in genes coding for IFs cause an array of different diseases, many related to stem cell dysfunction, but the molecular mechanisms remain unresolved. Here, we provide a comprehensive overview of how IFs interact with and regulate the activity, localization and function of different signaling proteins in stem cells, and how the assembly state and PTM profile of IFs may affect these processes. Identifying when, where and how IFs and cell signaling congregate, will expand our understanding of IF‐linked stem cell dysfunction during development and disease.
Collapse
Affiliation(s)
- Marika Sjöqvist
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Daniel Antfolk
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Freddy Suarez-Rodriguez
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland
| | - Cecilia Sahlgren
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Turku Bioscience, Åbo Akademi University and University of Turku, Turku, Finland.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
11
|
Chen X, Liang H, Xi Z, Yang Y, Shan H, Wang B, Zhong Z, Xu C, Yang GY, Sun Q, Sun Y, Bian L. BM-MSC Transplantation Alleviates Intracerebral Hemorrhage-Induced Brain Injury, Promotes Astrocytes Vimentin Expression, and Enhances Astrocytes Antioxidation via the Cx43/Nrf2/HO-1 Axis. Front Cell Dev Biol 2020; 8:302. [PMID: 32457903 PMCID: PMC7227447 DOI: 10.3389/fcell.2020.00302] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/07/2020] [Indexed: 12/30/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a particularly severe form of stroke, and reactive astrogliosis is a common response following injury to the central nervous system (CNS). Mesenchymal stem cells (MSCs) are reported to promote neurogenesis and alleviate the late side effects in injured brain regions. Gap junctions (Gjs) are abundant in the brain, where the richest connexin (Cx) is Cx43, most prominently expressed in astrocytes. Nuclear factor erythroid 2-related factor 2 (Nrf2) is an essential transcription factor regulating antioxidant reactions. Here, we aimed to explore whether bone marrow MSCs (BM-MSCs) could alleviate brain injury and protect astrocytes from apoptosis, by regulating Cx43 and Nrf2. We validated the effect of BM-MSC transplantation in an ICH model in vivo and in vitro and detected changes using immunofluorescence, as well as protein and mRNA expression of glial fibrillary acidic protein (GFAP), vimentin (VIM), Cx43, Nrf2, and heme oxygenase-1 (HO-1). Our results showed that BM-MSC transplantation attenuated brain injury after ICH and upregulated VIM expression in vivo and in vitro. Additionally, Cx43 upregulation and Nrf2 nuclear translocation were observed in astrocytes cocultured with BM-MSC. Knockdown of Cx43 by siRNA restrained Nrf2 nuclear translocation. Cx43 and Nrf2 had a connection as determined by immunofluorescence and coimmunoprecipitation. We demonstrated that astrocytes undergo astroglial-mesenchymal phenotype switching and have anti-apoptotic abilities after BM-MSC transplantation, where Cx43 upregulation triggers Nrf2 nuclear translocation and promotes its phase II enzyme expression. The Cx43/Nrf2 interaction of astrocytes after BM-MSC transplantation may provide an important therapeutic target in the management of ICH.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huaibin Liang
- Department of Neurology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiyu Xi
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Yang
- Department of Neurosurgery, Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huimin Shan
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Baofeng Wang
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhihong Zhong
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Canxin Xu
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qingfang Sun
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuhao Sun
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liuguan Bian
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
12
|
Chen X, Xu CX, Liang H, Xi Z, Pan J, Yang Y, Sun Q, Yang G, Sun Y, Bian L. Bone marrow mesenchymal stem cells transplantation alleviates brain injury after intracerebral hemorrhage in mice through the Hippo signaling pathway. Aging (Albany NY) 2020; 12:6306-6323. [PMID: 32271159 PMCID: PMC7185092 DOI: 10.18632/aging.103025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/02/2020] [Indexed: 01/10/2023]
Abstract
Intracerebral hemorrhage (ICH) is a common acute nervous system disease with high mortality and severe disability. Mesenchymal stem cells (MSCs) have been reported to promote neurogenesis and to alleviate side effects in areas of brain injury areas. The Hippo pathway regulates diverse cellular processes, including cell survival, proliferation, differentiation, and organ size. Here, we found that transplantation of bone marrow MSCs (BM-MSCs) into the brains of mice could alleviate ICH-mediated injury and protect astrocytes from apoptosis by regulating mammalian sterile 20-like kinase (MST)1 and Yes-associated protein (YAP). Knocking down of MST1 by si-RNA triggered YAP nuclear translocation. We further demonstrated that astrocytes undergo astroglial-mesenchymal phenotype switching and become capable of proliferating after BM-MSC transplantation via the Hippo signaling pathway. Together, our identification of the Hippo pathway in mediating the beneficial effects of BM-MSCs may provide a novel therapeutic target in the treatment and management of ICH.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Can-Xin Xu
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Huaibin Liang
- Department of Neurology, Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Zhiyu Xi
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jiaji Pan
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yong Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Qingfang Sun
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Guoyuan Yang
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yuhao Sun
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Liuguan Bian
- Department of Neurosurgery, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| |
Collapse
|
13
|
Strouhalova K, Přechová M, Gandalovičová A, Brábek J, Gregor M, Rosel D. Vimentin Intermediate Filaments as Potential Target for Cancer Treatment. Cancers (Basel) 2020; 12:E184. [PMID: 31940801 PMCID: PMC7017239 DOI: 10.3390/cancers12010184] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 02/06/2023] Open
Abstract
Intermediate filaments constitute the third component of the cellular skeleton. Unlike actin and microtubule cytoskeletons, the intermediate filaments are composed of a wide variety of structurally related proteins showing distinct expression patterns in tissues and cell types. Changes in the expression patterns of intermediate filaments are often associated with cancer progression; in particular with phenotypes leading to increased cellular migration and invasion. In this review we will describe the role of vimentin intermediate filaments in cancer cell migration, cell adhesion structures, and metastasis formation. The potential for targeting vimentin in cancer treatment and the development of drugs targeting vimentin will be reviewed.
Collapse
Affiliation(s)
- Katerina Strouhalova
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Magdalena Přechová
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Aneta Gandalovičová
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| | - Martin Gregor
- Laboratory of Integrative Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, 14220 Prague, Czech Republic;
| | - Daniel Rosel
- Department of Cell Biology, Charles University, Viničná 7, 12843 Prague, Czech Republic; (K.S.); (A.G.); (J.B.)
- Biotechnology and Biomedicine Centre of the Academy of Sciences and Charles University (BIOCEV), Průmyslová 595, 25242 Vestec u Prahy, Czech Republic
| |
Collapse
|
14
|
Radiosensitization and a Less Aggressive Phenotype of Human Malignant Glioma Cells Expressing Isocitrate Dehydrogenase 1 (IDH1) Mutant Protein: Dissecting the Mechanisms. Cancers (Basel) 2019; 11:cancers11060889. [PMID: 31242696 PMCID: PMC6627228 DOI: 10.3390/cancers11060889] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 01/02/2023] Open
Abstract
The presence of an isocitrate dehydrogenase 1 (IDH1) mutation is associated with a less aggressive phenotype, increased sensitivity to radiation, and increased overall survival in patients with diffuse glioma. Based on in vitro experimentations in malignant glioma cell lines, the consequences on cellular processes of IDH1R132H expression were analyzed. The results revealed that IDH1R132H expression enhanced the radiation induced accumulation of residual γH2AX foci and decreased the amount of glutathione (GSH) independent of the oxygen status. In addition, expression of the mutant IDH1 caused a significant increase of cell stiffness and induced an altered organization of the cytoskeleton, which has been shown to reinforce cell stiffness. Furthermore, IDH1R132H expression decreased the expression of vimentin, an important component of the cytoskeleton and regulator of the cell stiffness. The results emphasize the important role of mutant IDH1 in treatment of patients with diffuse gliomas especially in response to radiation. Hence, detection of the genetic status of IDH1 before therapy massively expands the utility of immunohistochemistry to accurately distinguish patients with a less aggressive and radiosensitive IDH1-mutant diffuse glioma suitable for radiotherapy from those with a more aggressive IDH1-wildtype diffuse glioma who might benefit from an individually intensified therapy comprising radiotherapy and alternative medical treatments.
Collapse
|
15
|
Sit B, Gutmann D, Iskratsch T. Costameres, dense plaques and podosomes: the cell matrix adhesions in cardiovascular mechanosensing. J Muscle Res Cell Motil 2019; 40:197-209. [PMID: 31214894 PMCID: PMC6726830 DOI: 10.1007/s10974-019-09529-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/15/2019] [Indexed: 12/12/2022]
Abstract
The stiffness of the cardiovascular environment changes during ageing and in disease and contributes to disease incidence and progression. For instance, increased arterial stiffness can lead to atherosclerosis, while stiffening of the heart due to fibrosis can increase the chances of heart failure. Cells can sense the stiffness of the extracellular matrix through integrin adhesions and other mechanosensitive structures and in response to this initiate mechanosignalling pathways that ultimately change the cellular behaviour. Over the past decades, interest in mechanobiology has steadily increased and with this also our understanding of the molecular basis of mechanosensing and transduction. However, much of our knowledge about the mechanisms is derived from studies investigating focal adhesions in non-muscle cells, which are distinct in several regards from the cell-matrix adhesions in cardiomyocytes (costameres) or vascular smooth muscle cells (dense plaques or podosomes). Therefore, we will look here first at the evidence for mechanical sensing in the cardiovascular system, before comparing the different cytoskeletal arrangements and adhesion sites in cardiomyocytes and vascular smooth muscle cells and what is known about mechanical sensing through the various structures.
Collapse
Affiliation(s)
- Brian Sit
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK
| | - Daniel Gutmann
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, London, UK.
| |
Collapse
|
16
|
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
|
17
|
The Cytoskeleton-A Complex Interacting Meshwork. Cells 2019; 8:cells8040362. [PMID: 31003495 PMCID: PMC6523135 DOI: 10.3390/cells8040362] [Citation(s) in RCA: 232] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/22/2022] Open
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
18
|
De Pascalis C, Pérez-González C, Seetharaman S, Boëda B, Vianay B, Burute M, Leduc C, Borghi N, Trepat X, Etienne-Manneville S. Intermediate filaments control collective migration by restricting traction forces and sustaining cell-cell contacts. J Cell Biol 2018; 217:3031-3044. [PMID: 29980627 PMCID: PMC6122997 DOI: 10.1083/jcb.201801162] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 05/13/2018] [Accepted: 06/12/2018] [Indexed: 11/22/2022] Open
Abstract
Using an in vitro wound healing assay, De Pascalis et al. show that intermediate filaments (IFs) participate in the dynamics of the acto-myosin network and its association with adhesions in astrocytes during collective migration. Glial IFs control the distribution of forces and the interactions between neighboring cells, ultimately determining the speed and direction of collective migration. Mesenchymal cell migration relies on the coordinated regulation of the actin and microtubule networks that participate in polarized cell protrusion, adhesion, and contraction. During collective migration, most of the traction forces are generated by the acto-myosin network linked to focal adhesions at the front of leader cells, which transmit these pulling forces to the followers. Here, using an in vitro wound healing assay to induce polarization and collective directed migration of primary astrocytes, we show that the intermediate filament (IF) network composed of vimentin, glial fibrillary acidic protein, and nestin contributes to directed collective movement by controlling the distribution of forces in the migrating cell monolayer. Together with the cytoskeletal linker plectin, these IFs control the organization and dynamics of the acto-myosin network, promoting the actin-driven treadmilling of adherens junctions, thereby facilitating the polarization of leader cells. Independently of their effect on adherens junctions, IFs influence the dynamics and localization of focal adhesions and limit their mechanical coupling to the acto-myosin network. We thus conclude that IFs promote collective directed migration in astrocytes by restricting the generation of traction forces to the front of leader cells, preventing aberrant tractions in the followers, and by contributing to the maintenance of lateral cell–cell interactions.
Collapse
Affiliation(s)
- Chiara De Pascalis
- Institut Pasteur Paris, Centre National de la Recherche Scientifique UMR3691, Cell Polarity, Migration, and Cancer Unit, Institut National de la Santé et de la Recherche Médicale, Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Sorbonne Universités, University Pierre and Marie Curie Université Paris 06, L'Institut de Formation Doctorale, Paris, France
| | - Carlos Pérez-González
- Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain.,Facultat de Medicina, University of Barcelona, Barcelona, Spain
| | - Shailaja Seetharaman
- Institut Pasteur Paris, Centre National de la Recherche Scientifique UMR3691, Cell Polarity, Migration, and Cancer Unit, Institut National de la Santé et de la Recherche Médicale, Equipe Labellisée Ligue Contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Batiste Boëda
- Institut Pasteur Paris, Centre National de la Recherche Scientifique UMR3691, Cell Polarity, Migration, and Cancer Unit, Institut National de la Santé et de la Recherche Médicale, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Benoit Vianay
- University of Paris Diderot, Institut National de la Santé et de la Recherche Médicale, Commissariat à l'Energie Atomique, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, Paris, France.,University of Grenoble-Alpes, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Biosciences and Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, CytoMorpho Lab, Grenoble, France
| | - Mithila Burute
- University of Paris Diderot, Institut National de la Santé et de la Recherche Médicale, Commissariat à l'Energie Atomique, Hôpital Saint Louis, Institut Universitaire d'Hematologie, UMRS1160, CytoMorpho Lab, Paris, France.,University of Grenoble-Alpes, Commissariat à l'Energie Atomique, Centre National de la Recherche Scientifique, Institut National de la Recherche Agronomique, Biosciences and Biotechnology Institute of Grenoble, Laboratoire de Physiologie Cellulaire et Végétale, CytoMorpho Lab, Grenoble, France
| | - Cécile Leduc
- Institut Pasteur Paris, Centre National de la Recherche Scientifique UMR3691, Cell Polarity, Migration, and Cancer Unit, Institut National de la Santé et de la Recherche Médicale, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Nicolas Borghi
- Institut Jacques Monod, Unité Mixe de Recherche 7592, Centre National de la Recherche Scientifique, Université Paris-Diderot, Paris, France
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain.,Facultat de Medicina, University of Barcelona, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina, Madrid, Spain
| | - Sandrine Etienne-Manneville
- Institut Pasteur Paris, Centre National de la Recherche Scientifique UMR3691, Cell Polarity, Migration, and Cancer Unit, Institut National de la Santé et de la Recherche Médicale, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| |
Collapse
|
19
|
Sanghvi-Shah R, Weber GF. Intermediate Filaments at the Junction of Mechanotransduction, Migration, and Development. Front Cell Dev Biol 2017; 5:81. [PMID: 28959689 PMCID: PMC5603733 DOI: 10.3389/fcell.2017.00081] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/30/2017] [Indexed: 01/04/2023] Open
Abstract
Mechanically induced signal transduction has an essential role in development. Cells actively transduce and respond to mechanical signals and their internal architecture must manage the associated forces while also being dynamically responsive. With unique assembly-disassembly dynamics and physical properties, cytoplasmic intermediate filaments play an important role in regulating cell shape and mechanical integrity. While this function has been recognized and appreciated for more than 30 years, continually emerging data also demonstrate important roles of intermediate filaments in cell signal transduction. In this review, with a particular focus on keratins and vimentin, the relationship between the physical state of intermediate filaments and their role in mechanotransduction signaling is illustrated through a survey of current literature. Association with adhesion receptors such as cadherins and integrins provides a critical interface through which intermediate filaments are exposed to forces from a cell's environment. As a consequence, these cytoskeletal networks are posttranslationally modified, remodeled and reorganized with direct impacts on local signal transduction events and cell migratory behaviors important to development. We propose that intermediate filaments provide an opportune platform for cells to both cope with mechanical forces and modulate signal transduction.
Collapse
Affiliation(s)
- Rucha Sanghvi-Shah
- Department of Biological Sciences, Rutgers University-NewarkNewark, NJ, United States
| | - Gregory F Weber
- Department of Biological Sciences, Rutgers University-NewarkNewark, NJ, United States
| |
Collapse
|
20
|
Cheng F, Eriksson JE. Intermediate Filaments and the Regulation of Cell Motility during Regeneration and Wound Healing. Cold Spring Harb Perspect Biol 2017; 9:9/9/a022046. [PMID: 28864602 DOI: 10.1101/cshperspect.a022046] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SUMMARYIntermediate filaments (IFs) comprise a diverse group of flexible cytoskeletal structures, the assembly, dynamics, and functions of which are regulated by posttranslational modifications. Characteristically, the expression of IF proteins is specific for tissues, differentiation stages, cell types, and functional contexts. Recent research has rapidly expanded the knowledge of IF protein functions. From being regarded as primarily structural proteins, it is now well established that IFs act as powerful modulators of cell motility and migration, playing crucial roles in wound healing and tissue regeneration, as well as inflammatory and immune responses. Although many of these IF-associated functions are essential for tissue repair, the involvement of IF proteins has been established in many additional facets of tissue healing and regeneration. Here, we review the recent progress in understanding the multiple functions of cytoplasmic IFs that relate to cell motility in the context of wound healing, taking examples from studies on keratin, vimentin, and nestin. Wound healing and regeneration include orchestration of a broad range of cellular processes, including regulation of cell attachment and migration, proliferation, differentiation, immune responses, angiogenesis, and remodeling of the extracellular matrix. In this respect, IF proteins now emerge as multifactorial and tissue-specific integrators of tissue regeneration, thereby acting as essential guardian biopolymers at the interface between health and disease, the failing of which contributes to a diverse range of pathologies.
Collapse
Affiliation(s)
- Fang Cheng
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, FI-20520 Turku, Finland.,Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, FI-20520, Turku, Finland
| | - John E Eriksson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, FI-20520 Turku, Finland.,Turku Centre for Biotechnology, Åbo Akademi University and University of Turku, FI-20520, Turku, Finland
| |
Collapse
|
21
|
Leduc C, Etienne-Manneville S. Regulation of microtubule-associated motors drives intermediate filament network polarization. J Cell Biol 2017; 216:1689-1703. [PMID: 28432079 PMCID: PMC5461013 DOI: 10.1083/jcb.201607045] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 01/16/2017] [Accepted: 03/03/2017] [Indexed: 11/22/2022] Open
Abstract
Intermediate filaments (IFs) are key players in the control of cell morphology and structure as well as in active processes such as cell polarization, migration, and mechanoresponses. However, the regulatory mechanisms controlling IF dynamics and organization in motile cells are still poorly understood. In this study, we investigate the mechanisms leading to the polarized rearrangement of the IF network along the polarity axis. Using photobleaching and photoconversion experiments in glial cells expressing vimentin, glial fibrillary acidic protein, and nestin, we show that the distribution of cytoplasmic IFs results from a continuous turnover based on the cooperation of an actin-dependent retrograde flow and anterograde and retrograde microtubule-dependent transports. During wound-induced astrocyte polarization, IF transport becomes directionally biased from the cell center toward the cell front. Such asymmetry in the transport is mainly caused by a Cdc42- and atypical PKC-dependent inhibition of dynein-dependent retrograde transport. Our results show how polarity signaling can affect the dynamic turnover of the IF network to promote the polarization of the network itself.
Collapse
Affiliation(s)
- Cécile Leduc
- Institut Pasteur Paris, Cell Polarity, Migration and Cancer Unit, UMR 3691, Equipe Labellisée Ligue Contre le Cancer, Centre National de la Recherché Scientifique, 75724 Paris, France
| | - Sandrine Etienne-Manneville
- Institut Pasteur Paris, Cell Polarity, Migration and Cancer Unit, UMR 3691, Equipe Labellisée Ligue Contre le Cancer, Centre National de la Recherché Scientifique, 75724 Paris, France.
| |
Collapse
|
22
|
Jones JCR, Kam CY, Harmon RM, Woychek AV, Hopkinson SB, Green KJ. Intermediate Filaments and the Plasma Membrane. Cold Spring Harb Perspect Biol 2017; 9:9/1/a025866. [PMID: 28049646 DOI: 10.1101/cshperspect.a025866] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A variety of intermediate filament (IF) types show intricate association with plasma membrane proteins, including receptors and adhesion molecules. The molecular basis of linkage of IFs to desmosomes at sites of cell-cell interaction and hemidesmosomes at sites of cell-matrix adhesion has been elucidated and involves IF-associated proteins. However, IFs also interact with focal adhesions and cell-surface molecules, including dystroglycan. Through such membrane interactions, it is well accepted that IFs play important roles in the establishment and maintenance of tissue integrity. However, by organizing cell-surface complexes, IFs likely regulate, albeit indirectly, signaling pathways that are key to tissue homeostasis and repair.
Collapse
Affiliation(s)
- Jonathan C R Jones
- The School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Chen Yuan Kam
- Departments of Dermatology and Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Robert M Harmon
- Departments of Dermatology and Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Alexandra V Woychek
- The School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Susan B Hopkinson
- The School of Molecular Biosciences, Washington State University, Pullman, Washington 99164
| | - Kathleen J Green
- Departments of Dermatology and Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|
23
|
Manninen A, Varjosalo M. A proteomics view on integrin-mediated adhesions. Proteomics 2016; 17. [PMID: 27723259 DOI: 10.1002/pmic.201600022] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/05/2016] [Accepted: 10/06/2016] [Indexed: 01/15/2023]
Abstract
Individual cells in multicellular organisms constantly explore their microenvironment, or niche, to obtain spatial information that is used to regulate cell behavior to maintain tissue integrity. The extracellular matrix (ECM) is an important source of such spatial information. Binding of the integrin family receptors to the ECM triggers formation of integrin adhesion complexes (IACs) that link the ECM network to cellular cytoskeleton via remarkably large multiprotein complexes collectively referred to as the integrin adhesome. Recent advances in proteomics have enabled researchers to study the IAC composition in detail. Various biochemical IAC isolation methods and culture conditions have been employed to study the composition and dynamics of integrin-mediated adhesions mainly in fibroblasts and lymphoblasts. These studies have led to identification of daunting lists of potential IAC components. This review focuses on the current status of proteomics-driven research seeking to understand integrin functions by comprehensive analysis of IAC components. These systems level approaches have revealed the complexity of biochemical and biomechanical signals that are processed at IACs and provide a novel insight into how these signals are conveyed to regulate cellular behavior.
Collapse
Affiliation(s)
- Aki Manninen
- Biocenter Oulu, Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, Biocenter 3, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Moeton M, Stassen OMJA, Sluijs JA, van der Meer VWN, Kluivers LJ, van Hoorn H, Schmidt T, Reits EAJ, van Strien ME, Hol EM. GFAP isoforms control intermediate filament network dynamics, cell morphology, and focal adhesions. Cell Mol Life Sci 2016; 73:4101-20. [PMID: 27141937 PMCID: PMC5043008 DOI: 10.1007/s00018-016-2239-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 04/12/2016] [Accepted: 04/21/2016] [Indexed: 11/01/2022]
Abstract
Glial fibrillary acidic protein (GFAP) is the characteristic intermediate filament (IF) protein in astrocytes. Expression of its main isoforms, GFAPα and GFAPδ, varies in astrocytes and astrocytoma implying a potential regulatory role in astrocyte physiology and pathology. An IF-network is a dynamic structure and has been functionally linked to cell motility, proliferation, and morphology. There is a constant exchange of IF-proteins with the network. To study differences in the dynamic properties of GFAPα and GFAPδ, we performed fluorescence recovery after photobleaching experiments on astrocytoma cells with fluorescently tagged GFAPs. Here, we show for the first time that the exchange of GFP-GFAPδ was significantly slower than the exchange of GFP-GFAPα with the IF-network. Furthermore, a collapsed IF-network, induced by GFAPδ expression, led to a further decrease in fluorescence recovery of both GFP-GFAPα and GFP-GFAPδ. This altered IF-network also changed cell morphology and the focal adhesion size, but did not alter cell migration or proliferation. Our study provides further insight into the modulation of the dynamic properties and functional consequences of the IF-network composition.
Collapse
Affiliation(s)
- Martina Moeton
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Oscar M J A Stassen
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Soft Tissue Biomechanics & Engineering, Department of biomedical engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Jacqueline A Sluijs
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Vincent W N van der Meer
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Liselot J Kluivers
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Hedde van Hoorn
- Physics of Life Processes, Leiden Institute of Physics, Leiden, The Netherlands
| | - Thomas Schmidt
- Physics of Life Processes, Leiden Institute of Physics, Leiden, The Netherlands
| | - Eric A J Reits
- Cell Biology and Histology, AMC Medical Center, Amsterdam, The Netherlands
| | - Miriam E van Strien
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Elly M Hol
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands.
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
25
|
Charrier EE, Asnacios A, Milloud R, De Mets R, Balland M, Delort F, Cardoso O, Vicart P, Batonnet-Pichon S, Hénon S. Desmin Mutation in the C-Terminal Domain Impairs Traction Force Generation in Myoblasts. Biophys J 2016; 110:470-480. [PMID: 26789769 DOI: 10.1016/j.bpj.2015.11.3518] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 11/06/2015] [Accepted: 11/23/2015] [Indexed: 02/08/2023] Open
Abstract
The cytoskeleton plays a key role in the ability of cells to both resist mechanical stress and generate force, but the precise involvement of intermediate filaments in these processes remains unclear. We focus here on desmin, a type III intermediate filament, which is specifically expressed in muscle cells and serves as a skeletal muscle differentiation marker. By using several complementary experimental techniques, we have investigated the impact of overexpressing desmin and expressing a mutant desmin on the passive and active mechanical properties of C2C12 myoblasts. We first show that the overexpression of wild-type-desmin increases the overall rigidity of the cells, whereas the expression of a mutated E413K desmin does not. This mutation in the desmin gene is one of those leading to desminopathies, a subgroup of myopathies associated with progressive muscular weakness that are characterized by the presence of desmin aggregates and a disorganization of sarcomeres. We show that the expression of this mutant desmin in C2C12 myoblasts induces desmin network disorganization, desmin aggregate formation, and a small decrease in the number and total length of stress fibers. We finally demonstrate that expression of the E413K mutant desmin also alters the traction forces generation of single myoblasts lacking organized sarcomeres.
Collapse
Affiliation(s)
- Elisabeth E Charrier
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR 8251, Paris, France; Matière et Systèmes Complexes, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR 7057, Paris, France
| | - Atef Asnacios
- Matière et Systèmes Complexes, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR 7057, Paris, France
| | - Rachel Milloud
- LIPhy Université Grenoble 1, CNRS, UMR 5588, Grenoble, France
| | - Richard De Mets
- LIPhy Université Grenoble 1, CNRS, UMR 5588, Grenoble, France
| | - Martial Balland
- LIPhy Université Grenoble 1, CNRS, UMR 5588, Grenoble, France
| | - Florence Delort
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR 8251, Paris, France
| | - Olivier Cardoso
- Matière et Systèmes Complexes, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR 7057, Paris, France
| | - Patrick Vicart
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR 8251, Paris, France
| | - Sabrina Batonnet-Pichon
- Unité de Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR 8251, Paris, France
| | - Sylvie Hénon
- Matière et Systèmes Complexes, Université Paris Diderot, Sorbonne Paris Cité, CNRS, UMR 7057, Paris, France.
| |
Collapse
|
26
|
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
|
27
|
Mendez MG, Restle D, Janmey PA. Vimentin enhances cell elastic behavior and protects against compressive stress. Biophys J 2015; 107:314-323. [PMID: 25028873 DOI: 10.1016/j.bpj.2014.04.050] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/04/2014] [Accepted: 04/30/2014] [Indexed: 02/05/2023] Open
Abstract
Vimentin intermediate filament expression is a hallmark of epithelial-to-mesenchymal transitions, and vimentin is involved in the maintenance of cell mechanical properties, cell motility, adhesion, and other signaling pathways. A common feature of vimentin-expressing cells is their routine exposure to mechanical stress. Intermediate filaments are unique among cytoskeletal polymers in resisting large deformations in vitro, yet vimentin's mechanical role in the cell is not clearly understood. We use atomic force microscopy to compare the viscoelastic properties of normal and vimentin-null (vim(-/-)) mouse embryo fibroblasts (mEFs) on substrates of different stiffnesses, spread to different areas, and subjected to different compression patterns. In minimally perturbed mEF, vimentin contributes little to the elastic modulus at any indentation depth in cells spread to average areas. On a hard substrate however, the elastic moduli of maximally spread mEFs are greater than those of vim(-/-)mEF. Comparison of the plastic deformation resulting from controlled compression of the cell cortex shows that vimentin's enhancement of elastic behavior increases with substrate stiffness. The elastic moduli of normal mEFs are more stable over time than those of vim(-/-)mEFs when cells are subject to ongoing oscillatory compression, particularly on a soft substrate. In contrast, increasing compressive strain over time shows a greater role for vimentin on a hard substrate. Under both conditions, vim(-/-)mEFs exhibit more variable responses, indicating a loss of regulation. Finally, normal mEFs are more contractile in three-dimensional collagen gels when seeded at low density, when cell-matrix contacts dominate, whereas contractility of vim(-/-)mEF is greater at higher densities when cell-cell contacts are abundant. Addition of fibronectin to gel constructs equalizes the contractility of the two cell types. These results show that the Young's moduli of normal and vim(-/-)mEFs are substrate stiffness dependent even when the spread area is similar, and that vimentin protects against compressive stress and preserves mechanical integrity by enhancing cell elastic behavior.
Collapse
Affiliation(s)
- M G Mendez
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - D Restle
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania
| | - P A Janmey
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
28
|
Bargagna-Mohan P, Lei L, Thompson A, Shaw C, Kasahara K, Inagaki M, Mohan R. Vimentin Phosphorylation Underlies Myofibroblast Sensitivity to Withaferin A In Vitro and during Corneal Fibrosis. PLoS One 2015; 10:e0133399. [PMID: 26186445 PMCID: PMC4506086 DOI: 10.1371/journal.pone.0133399] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 06/26/2015] [Indexed: 12/21/2022] Open
Abstract
Vimentin is a newly recognized target for corneal fibrosis. Using primary rabbit corneal fibroblasts and myofibroblasts we show that myofibroblasts, unlike fibroblasts, display impaired cell spreading and cell polarization, which is associated with increased levels of soluble serine-38 phosphorylated vimentin (pSer38Vim). This pSer38Vim isoform is inefficiently incorporated into growing vimentin intermediate filaments (IFs) of myofibroblasts during cell spreading, and as a result, myofibroblasts maintain higher soluble pSer38Vim levels compared to fibroblasts. Moreover, the soluble vimentin-targeting small molecule and fibrotic inhibitor withaferin A (WFA) causes a potent blockade of cell spreading selectively in myofibroblasts by targeting soluble pSer38Vim for hyperphosphorylation. WFA treatment does not induce vimentin hyperphosphorylation in fibroblasts. This hyperphosphorylated pSer38Vim species in WFA-treated myofibroblasts becomes complexed with adaptor protein filamin A (FlnA), and these complexes appear as short squiggles when displaced from focal adhesions. The extracellular-signal regulated kinase (ERK) is also phosphorylated (pERK) in response to WFA, but surprisingly, pERK does not enter the nucleus but remains bound to pSer38Vim in cytoplasmic complexes. Using a model of corneal alkali injury, we show that fibrotic corneas of wild type mice possess high levels of pERK, whereas injured corneas of vimentin-deficient (Vim KO) mice that heal with reduced fibrosis have highly reduced pERK expression. Finally, WFA treatment causes a decrease in pERK and pSer38Vim expression in healing corneas of wild type mice. Taken together, these findings identify a hereto-unappreciated role for pSer38Vim as an important determinant of myofibroblast sensitivity to WFA.
Collapse
Affiliation(s)
- Paola Bargagna-Mohan
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Ling Lei
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Alexis Thompson
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Camille Shaw
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Kousuke Kasahara
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Masaki Inagaki
- Division of Biochemistry, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Royce Mohan
- From the Department of Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
29
|
Zaidel-Bar R, Zhenhuan G, Luxenburg C. The contractome – a systems view of actomyosin contractility in non-muscle cells. J Cell Sci 2015; 128:2209-17. [DOI: 10.1242/jcs.170068] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/27/2015] [Indexed: 12/21/2022] Open
Abstract
ABSTRACT
Actomyosin contractility is a highly regulated process that affects many fundamental biological processes in each and every cell in our body. In this Cell Science at a Glance article and the accompanying poster, we mined the literature and databases to map the contractome of non-muscle cells. Actomyosin contractility is involved in at least 49 distinct cellular functions that range from providing cell architecture to signal transduction and nuclear activity. Containing over 100 scaffolding and regulatory proteins, the contractome forms a highly complex network with more than 230 direct interactions between its components, 86 of them involving phosphorylation. Mapping these interactions, we identify the key regulatory pathways involved in the assembly of actomyosin structures and in activating myosin to produce contractile forces within non-muscle cells at the exact time and place necessary for cellular function.
Collapse
Affiliation(s)
- Ronen Zaidel-Bar
- Mechanobiology Institute, National University of Singapore, T-lab building #05-01, 5A Engineering Drive 1, 117411, Singapore
| | - Guo Zhenhuan
- Mechanobiology Institute, National University of Singapore, T-lab building #05-01, 5A Engineering Drive 1, 117411, Singapore
| | - Chen Luxenburg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, P.O. Box 39040, Tel Aviv 69978, Israel
| |
Collapse
|
30
|
Leduc C, Etienne-Manneville S. Intermediate filaments in cell migration and invasion: the unusual suspects. Curr Opin Cell Biol 2015; 32:102-12. [PMID: 25660489 DOI: 10.1016/j.ceb.2015.01.005] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Revised: 01/15/2015] [Accepted: 01/21/2015] [Indexed: 12/22/2022]
Abstract
Cell migration is a multistep process which relies on the coordination of cytoskeletal structures in space and time. While the roles of actin and microtubules have been investigated in great details, the lack of inhibitors and visualizing tools and the large number of proteins forming intermediate filaments (IFs) have delayed the characterization of IF functions during migration. However, a large body of evidence has progressively pointed to changes in IF composition as an important parameter in the regulation of cell migratory properties both during development and tumor invasion. More recent in-depth analyses show that IFs are dynamically reorganized to participate, together with microfilaments and microtubules, to the key steps leading to cell migration.
Collapse
Affiliation(s)
- Cécile Leduc
- Institut Pasteur - CNRS UMR 3691, Cell Polarity, Migration and Cancer Unit, 25 rue du Dr Roux, 75724 Paris Cedex 15, France
| | - Sandrine Etienne-Manneville
- Institut Pasteur - CNRS UMR 3691, Cell Polarity, Migration and Cancer Unit, 25 rue du Dr Roux, 75724 Paris Cedex 15, France.
| |
Collapse
|
31
|
Huber F, Boire A, López MP, Koenderink GH. Cytoskeletal crosstalk: when three different personalities team up. Curr Opin Cell Biol 2015; 32:39-47. [DOI: 10.1016/j.ceb.2014.10.005] [Citation(s) in RCA: 187] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/20/2014] [Accepted: 10/22/2014] [Indexed: 12/29/2022]
|
32
|
Iskratsch T, Wolfenson H, Sheetz MP. Appreciating force and shape—the rise of mechanotransduction in cell biology. Nat Rev Mol Cell Biol 2014; 15:825-33. [PMID: 25355507 PMCID: PMC9339222 DOI: 10.1038/nrm3903] [Citation(s) in RCA: 563] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although the shapes of organisms are encoded in their genome, the developmental processes that lead to the final form of vertebrates involve a constant feedback between dynamic mechanical forces, and cell growth and motility. Mechanobiology has emerged as a discipline dedicated to the study of the effects of mechanical forces and geometry on cell growth and motility—for example, during cell-matrix adhesion development—through the signalling process of mechanotransduction.
Collapse
Affiliation(s)
- Thomas Iskratsch
- 1] Department of Biological Sciences, Columbia University, New York 10027, USA. [2]
| | - Haguy Wolfenson
- 1] Department of Biological Sciences, Columbia University, New York 10027, USA. [2]
| | - Michael P Sheetz
- Mechanobiology Institute, National University of Singapore, Singapore 117411, and the Department of Biological Sciences, Columbia University, New York 10027, USA
| |
Collapse
|
33
|
Gruenbaum Y, Aebi U. Intermediate filaments: a dynamic network that controls cell mechanics. F1000PRIME REPORTS 2014; 6:54. [PMID: 25184044 PMCID: PMC4108948 DOI: 10.12703/p6-54] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In humans the superfamily of intermediate filament (IF) proteins is encoded by more than 70 different genes, which are expressed in a cell- and tissue-specific manner. IFs assemble into approximately 10 nm-wide filaments that account for the principal structural elements at the nuclear periphery, nucleoplasm, and cytoplasm. They are also required for organizing the microtubule and microfilament networks. In this review, we focus on the dynamics of IFs and how modifications regulate it. We also discuss the role of nuclear IF organization in determining nuclear mechanics as well as that of cytoplasmic IFs organization in maintaining cell stiffness, formation of lamellipodia, regulation of cell migration, and permitting cell adhesion.
Collapse
Affiliation(s)
- Yosef Gruenbaum
- Department of Genetics, Institute of Life Sciences, Hebrew University of JerusalemGivat Ram, Jerusalem 91904Israel
| | - Ueli Aebi
- Biozentrum, University of BaselKlingelbergerstrasse 70, CH-4056 BaselSwitzerland
| |
Collapse
|
34
|
Granato DC, Zanetti MR, Kawahara R, Yokoo S, Domingues RR, Aragão AZ, Agostini M, Carazzolle MF, Vidal RO, Flores IL, Korvala J, Cervigne NK, Silva ARS, Coletta RD, Graner E, Sherman NE, Leme AFP. Integrated proteomics identified up-regulated focal adhesion-mediated proteins in human squamous cell carcinoma in an orthotopic murine model. PLoS One 2014; 9:e98208. [PMID: 24858105 PMCID: PMC4032327 DOI: 10.1371/journal.pone.0098208] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 04/30/2014] [Indexed: 01/20/2023] Open
Abstract
Understanding the molecular mechanisms of oral carcinogenesis will yield important advances in diagnostics, prognostics, effective treatment, and outcome of oral cancer. Hence, in this study we have investigated the proteomic and peptidomic profiles by combining an orthotopic murine model of oral squamous cell carcinoma (OSCC), mass spectrometry-based proteomics and biological network analysis. Our results indicated the up-regulation of proteins involved in actin cytoskeleton organization and cell-cell junction assembly events and their expression was validated in human OSCC tissues. In addition, the functional relevance of talin-1 in OSCC adhesion, migration and invasion was demonstrated. Taken together, this study identified specific processes deregulated in oral cancer and provided novel refined OSCC-targeting molecules.
Collapse
Affiliation(s)
- Daniela C. Granato
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | - Mariana R. Zanetti
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | - Rebeca Kawahara
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | - Sami Yokoo
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | - Romênia R. Domingues
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | - Annelize Z. Aragão
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | - Michelle Agostini
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, Brazil
- Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Brazil
| | - Marcelo F. Carazzolle
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | - Ramon O. Vidal
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
| | - Isadora L. Flores
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, Brazil
| | | | - Nilva K. Cervigne
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, Brazil
| | - Alan R. S. Silva
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, Brazil
| | - Ricardo D. Coletta
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, Brazil
| | - Edgard Graner
- Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas, UNICAMP, Piracicaba, Brazil
| | - Nicholas E. Sherman
- W. M. Keck Biomedical Mass Spectrometry Lab. University of Virginia, Charlottesville, Virginia, United States of America
| | - Adriana F. Paes Leme
- Laboratório de Espectrometria de Massas, Laboratório Nacional de Biociências, LNBio, CNPEM, Campinas, Brazil
- * E-mail:
| |
Collapse
|
35
|
Ciobanasu C, Faivre B, Le Clainche C. Integrating actin dynamics, mechanotransduction and integrin activation: the multiple functions of actin binding proteins in focal adhesions. Eur J Cell Biol 2013; 92:339-48. [PMID: 24252517 DOI: 10.1016/j.ejcb.2013.10.009] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/10/2013] [Accepted: 10/23/2013] [Indexed: 12/16/2022] Open
Abstract
Focal adhesions are clusters of integrin transmembrane receptors that mechanically couple the extracellular matrix to the actin cytoskeleton during cell migration. Focal adhesions sense and respond to variations in force transmission along a chain of protein-protein interactions linking successively actin filaments, actin binding proteins, integrins and the extracellular matrix to adapt cell-matrix adhesion to the composition and mechanical properties of the extracellular matrix. This review focuses on the molecular mechanisms by which actin binding proteins integrate actin dynamics, mechanotransduction and integrin activation to control force transmission in focal adhesions.
Collapse
Affiliation(s)
- Corina Ciobanasu
- Laboratoire d'Enzymologie et Biochimie Structurales CNRS, avenue de la terrasse, 91198 Gif-sur-Yvette, France
| | | | | |
Collapse
|
36
|
Khotskaya YB, Beck BH, Hurst DR, Han Z, Xia W, Hung MC, Welch DR. Expression of metastasis suppressor BRMS1 in breast cancer cells results in a marked delay in cellular adhesion to matrix. Mol Carcinog 2013; 53:1011-26. [PMID: 24000122 DOI: 10.1002/mc.22068] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/22/2013] [Accepted: 06/17/2013] [Indexed: 12/29/2022]
Abstract
Metastatic dissemination is a multi-step process that depends on cancer cells' ability to respond to microenvironmental cues by adapting adhesion abilities and undergoing cytoskeletal rearrangement. Breast Cancer Metastasis Suppressor 1 (BRMS1) affects several steps of the metastatic cascade: it decreases survival in circulation, increases susceptibility to anoikis, and reduces capacity to colonize secondary organs. In this report, BRMS1 expression is shown to not significantly alter expression levels of integrin monomers, while time-lapse and confocal microscopy revealed that BRMS1-expressing cells exhibited reduced activation of both β1 integrin and focal adhesion kinase, and decreased localization of these molecules to sites of focal adhesions. Short-term plating of BRMS1-expressing cells onto collagen or fibronectin markedly decreased cytoskeletal reorganization and formation of cellular adhesion projections. Under 3D culture conditions, BRMS1-expressing cells remained rounded and failed to reorganize their cytoskeleton and form invasive colonies. Taken together, BRMS1-expressing breast cancer cells are greatly attenuated in their ability to respond to microenvironment changes. © 2013 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yekaterina B Khotskaya
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | | |
Collapse
|
37
|
Bargagna-Mohan P, Deokule SP, Thompson K, Wizeman J, Srinivasan C, Vooturi S, Kompella UB, Mohan R. Withaferin A effectively targets soluble vimentin in the glaucoma filtration surgical model of fibrosis. PLoS One 2013; 8:e63881. [PMID: 23667686 PMCID: PMC3648549 DOI: 10.1371/journal.pone.0063881] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 04/09/2013] [Indexed: 01/23/2023] Open
Abstract
Withaferin A (WFA) is a natural product that binds to soluble forms of the type III intermediate filament (IF) vimentin. Currently, it is unknown under what pathophysiological contexts vimentin is druggable, as cytoskeltal vimentin-IFs are abundantly expressed. To investigate druggability of vimentin, we exploited rabbit Tenon's capsule fibroblast (RbTCF) cell cultures and the rabbit glaucoma filtration surgical (GFS) model of fibrosis. WFA potently caused G₀/G₁ cell cycle inhibition (IC₅₀ 25 nM) in RbTCFs, downregulating ubiquitin E3 ligase skp2 and inducing p27(Kip1) expression. Transforming growth factor (TGF)-ß-induced myofibroblast transformation caused development of cell spheroids with numerous elongated invadopodia, which WFA blocked potently by downregulating soluble vimentin and α-smooth muscle actin (SMA) expression. In the pilot proof-of-concept study using the GFS model, subconjunctival injections of a low WFA dose reduced skp2 expression in Tenon's capsule and increased p27(Kip1) expression without significant alteration to vimentin-IFs. This treatment maintains significant nanomolar WFA concentrations in anterior segment tissues that correspond to WFA's cell cycle targeting activity. A ten-fold higher WFA dose caused potent downregulation of soluble vimentin and skp2 expression, but as found in cell cultures, no further increase in p27(Kip1) expression was observed. Instead, this high WFA dose potently induced vimentin-IF disruption and downregulated α-SMA expression that mimicked WFA activity in TGF-ß-treated RbTCFs that blocked cell contractile activity at submicromolar concentrations. These findings illuminate that localized WFA injection to ocular tissues exerts pharmacological control over the skp2-p27(Kip1) pathway by targeting of soluble vimentin in a model of surgical fibrosis.
Collapse
Affiliation(s)
- Paola Bargagna-Mohan
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Sunil P. Deokule
- Ophthalmology and Visual Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - Kyle Thompson
- Ophthalmology and Visual Science, University of Kentucky, Lexington, Kentucky, United States of America
| | - John Wizeman
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Cidambi Srinivasan
- Statistics, University of Kentucky, Lexington, Kentucky, United States of America
| | - Sunil Vooturi
- Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Uday B. Kompella
- Pharmaceutical Sciences, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Royce Mohan
- Neuroscience, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|