1
|
Yang Y, Akdemir AR, Rashik RA, Shihadeh Khater OA, Weng Z, Wang L, Zhong Y, Gallant ND. Guided neural stem cell differentiation by dynamic loading of 3D printed elastomeric scaffolds. J Mech Behav Biomed Mater 2025; 165:106940. [PMID: 39955829 DOI: 10.1016/j.jmbbm.2025.106940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/22/2025] [Accepted: 02/09/2025] [Indexed: 02/18/2025]
Abstract
The limited regenerative ability of "permanent" cells is a major barrier to treating conditions like spinal cord injury (SCI) and myocardial infarction (MI). The delivery of stem cells, which can generate various cell types, offer potential for personalized therapy with reduced immunoreaction and recovery time. However, restoring function to these tissues also requires new or replacement cells to align properly. Neurons, for example, must organize and extend parallel axons, mimicking their natural structure for directional signal propagation. Current stem cell differentiation methods lack guidance, resulting in randomly distributed axons and limited repair effectiveness. Advancing methods and materials to guide stem cell differentiation into functional, aligned nerve bundles is crucial for improving SCI treatment outcomes. This study aimed to develop an in vitro system to promote aligned neural differentiation by applying cyclic uniaxial tension to PC-12 stem cells adhered to 3D-printed elastic scaffolds. We created a simple loading device which can apply cyclic and controllable stretching force to a scaffold, which in turn transmits uniaxial tension to cells adhered to the scaffold during their differentiation. An elastomer ink for 3D printing scaffolds was formulated and surface treatment processes were investigated to enhance the cell-scaffold adhesion to support the dynamic loading. It was revealed that a corona discharge treatment while the scaffold is soaked with type I collagen can significantly enhance cell adhesion. A range of strain magnitudes and frequencies were revealed to enhance the differentiation of neural tissue derived PC-12 cells to neuron cells and increase the length of their neurites up to 76%. The combination of 3% maximum strain and 1 Hz loading frequency maximized differentiation and neurite extension. These findings demonstrate that dynamic mechanical stimulation enhances neural differentiation and organization, offering an alternative approach for regenerative therapies targeting SCI and similar conditions.
Collapse
Affiliation(s)
- Yi Yang
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Abdullah Revaha Akdemir
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Rafsan Ahmed Rashik
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Omar Ahmad Shihadeh Khater
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Zijian Weng
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA
| | - Long Wang
- Department of Civil and Environmental Engineering, California Polytechnic State University, San Luis Obispo, CA, 93407, USA
| | - Ying Zhong
- Sauvage Laboratory for Smart Materials, School of Integrated Circuits, Harbin Institute of Technology (Shenzhen), University Town of Shenzhen, Shenzhen, Guangdong, 518055, China.
| | - Nathan D Gallant
- Department of Mechanical Engineering, University of South Florida, 4202 E. Fowler Ave, Tampa, FL, 33620, USA.
| |
Collapse
|
2
|
Blatkiewicz M, Białas P, Taryma-Leśniak O, Mazgaj S, Hukowska-Szematowicz B, Jankowska A. Exploring vimentin expression and its protein interactors across diverse cancer types via the cancer genome atlas datasets: a comprehensive analysis. Rep Pract Oncol Radiother 2025; 30:88-99. [PMID: 40242413 PMCID: PMC11999022 DOI: 10.5603/rpor.104142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/19/2024] [Indexed: 04/18/2025] Open
Abstract
Background The global burden of cancer is escalating, with millions of individuals diagnosed and succumbing to the disease each year. Early detection is crucial for improving patient outcomes, yet many cancers are identified at advanced stages. Vimentin (VIM) has emerged as a promising biomarker with significant diagnostic and prognostic potential. Materials and methods This study investigates VIM expression and promoter methylation across various cancers using The Cancer Genome Atlas (TCGA) datasets. Additionally, we analyze protein-protein interactions and mutation frequencies using advanced bioinformatics tools. Results Our findings reveal that VIM is overexpressed in seven cancer types, including cholangiocarcinoma, glioblastoma multiforme, and breast invasive carcinoma. Notably, VIM expression is correlated with promoter methylation in specific cancers. Furthermore, we identify complex protein interactions involving VIM, highlighting its role in critical cellular processes such as proliferation and apoptosis. Conclusion These insights emphasize Vimentin's multifaceted role in cancer, suggesting its potential as both a therapeutic target and a diagnostic marker.
Collapse
Affiliation(s)
- Małgorzata Blatkiewicz
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznań, Poland
| | - Piotr Białas
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Poznań, Poland
| | - Olga Taryma-Leśniak
- Independent Clinical Epigenetics Laboratory, Pomeranian Medical University, Szczecin, Poland
| | - Szymon Mazgaj
- Institute of Biology, University of Szczecin, Szczecin, Poland
- Department of Blood Preparation, Regional Center for Blood Donation and Blood Treatment in Zielona Góra, Zielona Góra, Poland
| | - Beata Hukowska-Szematowicz
- Institute of Biology, University of Szczecin, Szczecin, Poland
- Molecular Biology and Biotechnology Center, University of Szczecin, Szczecin, Poland
| | - Anna Jankowska
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
3
|
Isert L, Passi M, Freystetter B, Grab M, Roidl A, Müller C, Mehta A, Sundararaghavan HG, Zahler S, Merkel OM. Cellular EMT-status governs contact guidance in an electrospun TACS-mimicking in vitro model. Mater Today Bio 2025; 30:101401. [PMID: 39759848 PMCID: PMC11699613 DOI: 10.1016/j.mtbio.2024.101401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
In this study, an advanced nanofiber breast cancer in vitro model was developed and systematically characterized including physico-chemical, cell-biological and biophysical parameters. Using electrospinning, the architecture of tumor-associated collagen signatures (TACS5 and TACS6) was mimicked. By employing a rotating cylinder or static plate collector set-up, aligned fibers (TACS5-like structures) and randomly orientated fibers (TACS6-like structures) fibers were produced, respectively. The biocompatibility of these fibers was enhanced by collagen coating, ensuring minimal toxicity and improved cell attachment. Various breast cancer cell lines (MCF7, HCC1954, MDA-MB-468, and MDA-MB-231) were cultured on these fibers to assess epithelial-to-mesenchymal transition (EMT) markers, cellular morphology, and migration. Aligned fibers (TACS5) significantly influenced EMT-related changes, promoting cellular alignment, spindle-shaped morphology and a highly migratory phenotype in mesenchymal and hybrid EMT cells (MDA-MB-468, MDA-MB-231). Conversely, epithelial cells (MCF7, HCC1954) showed limited response, but - under growth factor treatment - started to infiltrate the fibrous scaffold and underwent EMT-like changes, particularly on TACS5-mimicks, emphasizing the interplay of topographical cues and EMT induction. The biophysical analysis revealed a clear correlation between cellular EMT status and cell mechanics, with increased EMT correlating to decreased total cellular stiffness. Cancer cell mechanics, however, were found to be dynamic during biochemical and topographical EMT-induction, exceeding initial stiffness by up to 2-fold. These findings highlight the potential of TACS5-like nanofiber scaffolds in modeling the tumor microenvironment and studying cancer cell behavior and mechanics.
Collapse
Affiliation(s)
- Lorenz Isert
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| | - Mehak Passi
- Pharmaceutical Biology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Benedikt Freystetter
- Department of Cardiac Surgery, Ludwig Maximilians University München, Munich, Germany
| | - Maximilian Grab
- Department of Cardiac Surgery, Ludwig Maximilians University München, Munich, Germany
| | - Andreas Roidl
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christoph Müller
- Center of Drug Research, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Aditi Mehta
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| | | | - Stefan Zahler
- Pharmaceutical Biology, Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Olivia M. Merkel
- Pharmaceutical Technology and Biopharmaceutics, Department of Pharmacy, Ludwig-Maximilians-University München, Munich, Germany
| |
Collapse
|
4
|
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
|
5
|
Lefèvre C, Cook GM, Dinan AM, Torii S, Stewart H, Gibbons G, Nicholson AS, Echavarría-Consuegra L, Meredith LW, Lulla V, McGovern N, Kenyon JC, Goodfellow I, Deane JE, Graham SC, Lakatos A, Lambrechts L, Brierley I, Irigoyen N. Zika viruses encode 5' upstream open reading frames affecting infection of human brain cells. Nat Commun 2024; 15:8822. [PMID: 39394194 PMCID: PMC11470053 DOI: 10.1038/s41467-024-53085-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 09/30/2024] [Indexed: 10/13/2024] Open
Abstract
Zika virus (ZIKV), an emerging mosquito-borne flavivirus, is associated with congenital neurological complications. Here, we investigate potential pathological correlates of virus gene expression in representative ZIKV strains through RNA sequencing and ribosome profiling. In addition to the single long polyprotein found in all flaviviruses, we identify the translation of unrecognised upstream open reading frames (uORFs) in the genomic 5' region. In Asian/American strains, ribosomes translate uORF1 and uORF2, whereas in African strains, the two uORFs are fused into one (African uORF). We use reverse genetics to examine the impact on ZIKV fitness of different uORFs mutant viruses. We find that expression of the African uORF and the Asian/American uORF1 modulates virus growth and tropism in human cortical neurons and cerebral organoids, suggesting a potential role in neurotropism. Although the uORFs are expressed in mosquito cells, we do not see a measurable effect on transmission by the mosquito vector in vivo. The discovery of ZIKV uORFs sheds new light on the infection of the human brain cells by this virus and raises the question of their existence in other neurotropic flaviviruses.
Collapse
Affiliation(s)
- Charlotte Lefèvre
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Georgia M Cook
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Adam M Dinan
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Shiho Torii
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Hazel Stewart
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - George Gibbons
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
| | - Alex S Nicholson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | | | - Luke W Meredith
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Valeria Lulla
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Naomi McGovern
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Julia C Kenyon
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Janet E Deane
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Stephen C Graham
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - András Lakatos
- John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, Cambridge, UK
| | - Louis Lambrechts
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Insect-Virus Interactions Unit, Paris, France
| | - Ian Brierley
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
6
|
Mamalakis M, Macfarlane SC, Notley SV, Gad AKB, Panoutsos G. A novel pipeline employing deep multi-attention channels network for the autonomous detection of metastasizing cells through fluorescence microscopy. Comput Biol Med 2024; 181:109052. [PMID: 39216406 DOI: 10.1016/j.compbiomed.2024.109052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 08/09/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Metastasis driven by cancer cell migration is the leading cause of cancer-related deaths. It involves significant changes in the organization of the cytoskeleton, which includes the actin microfilaments and the vimentin intermediate filaments. Understanding how these filament change cells from normal to invasive offers insights that can be used to improve cancer diagnosis and therapy. We have developed a computational, transparent, large-scale and imaging-based pipeline, that can distinguish between normal human cells and their isogenically matched, oncogenically transformed, invasive and metastasizing counterparts, based on the spatial organization of actin and vimentin filaments in the cell cytoplasm. Due to the intricacy of these subcellular structures, manual annotation is not trivial to automate. We used established deep learning methods and our new multi-attention channel architecture. To ensure a high level of interpretability of the network, which is crucial for the application area, we developed an interpretable global explainable approach correlating the weighted geometric mean of the total cell images and their local GradCam scores. The methods offer detailed, objective and measurable understanding of how different components of the cytoskeleton contribute to metastasis, insights that can be used for future development of novel diagnostic tools, such as a nanometer level, vimentin filament-based biomarker for digital pathology, and for new treatments that significantly can increase patient survival.
Collapse
Affiliation(s)
- Michail Mamalakis
- School of Electrical and Electronic Engineering, University of Sheffield, Sheffield, UK; Insigneo Institute for in-silico, Medicine, University of Sheffield, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, and Department of Computer science, Sheffield, UK; Department of Psychiatry, Cambridge University, Cambridge, UK.
| | - Sarah C Macfarlane
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, UK
| | - Scott V Notley
- Insigneo Institute for in-silico, Medicine, University of Sheffield, Sheffield, UK; Department of Automatic Control and Systems Engineering, University of Sheffield, Sheffield, UK
| | - Annica K B Gad
- Insigneo Institute for in-silico, Medicine, University of Sheffield, Sheffield, UK; Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, UK; Madeira Chemistry Research Centre, University of Madeira, Funchal, Portugal; Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - George Panoutsos
- School of Electrical and Electronic Engineering, University of Sheffield, Sheffield, UK; Insigneo Institute for in-silico, Medicine, University of Sheffield, Sheffield, UK; Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, UK.
| |
Collapse
|
7
|
Pradeau-Phélut L, Etienne-Manneville S. Cytoskeletal crosstalk: A focus on intermediate filaments. Curr Opin Cell Biol 2024; 87:102325. [PMID: 38359728 DOI: 10.1016/j.ceb.2024.102325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 02/17/2024]
Abstract
The cytoskeleton, comprising actin microfilaments, microtubules, and intermediate filaments, is crucial for cell motility and tissue integrity. While prior studies largely focused on individual cytoskeletal networks, recent research underscores the interconnected nature of these systems in fundamental cellular functions like adhesion, migration, and division. Understanding the coordination of these distinct networks in both time and space is essential. This review synthesizes current findings on the intricate interplay between these networks, emphasizing the pivotal role of intermediate filaments. Notably, these filaments engage in extensive crosstalk with microfilaments and microtubules through direct molecular interactions, cytoskeletal linkers, and molecular motors that form molecular bridges, as well as via more complex regulation of intracellular signaling.
Collapse
Affiliation(s)
- Lucas Pradeau-Phélut
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur - CNRS UMR 3691, Université Paris-Cité, Équipe Labellisée Ligue Nationale Contre le Cancer 2023, 25 rue du Docteur Roux, F-75015, Paris, France; Sorbonne Université, Collège Doctoral, 4 place Jussieu, F-75005 Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur - CNRS UMR 3691, Université Paris-Cité, Équipe Labellisée Ligue Nationale Contre le Cancer 2023, 25 rue du Docteur Roux, F-75015, Paris, France.
| |
Collapse
|
8
|
Wang B, Dong J, Yang F, Ju T, Wang J, Qu K, Wang Y, Tian Y, Wang Z. Determining the degree of chromosomal instability in breast cancer cells by atomic force microscopy. Analyst 2024; 149:1988-1997. [PMID: 38420857 DOI: 10.1039/d3an01815f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Chromosomal instability (CIN) is a source of genetic variation and is highly linked to the malignance of cancer. Determining the degree of CIN is necessary for understanding the role that it plays in tumor development. There is currently a lack of research on high-resolution characterization of CIN and the relationship between CIN and cell mechanics. Here, a method to determine CIN of breast cancer cells by high resolution imaging with atomic force microscopy (AFM) is explored. The numerical and structural changes of chromosomes in human breast cells (MCF-10A), moderately malignant breast cells (MCF-7) and highly malignant breast cells (MDA-MB-231) were observed and analyzed by AFM. Meanwhile, the nuclei, cytoskeleton and cell mechanics of the three kinds of cells were also investigated. The results showed the differences in CIN between the benign and cancer cells. Also, the degree of structural CIN increased with enhanced malignancy of cancer cells. This was also demonstrated by calculating the probability of micronucleus formation in these three kinds of cells. Meanwhile, we found that the area of the nucleus was related to the number of chromosomes in the nucleus. In addition, reduced or even aggregated actin fibers led to decreased elasticities in MCF-7 and MDA-MB-231 cells. It was found that the rearrangement of actin fibers would affect the nucleus, and then lead to wrong mitosis and CIN. Using AFM to detect chromosomal changes in cells with different malignancy degrees provides a new detection method for the study of cell carcinogenesis with a perspective for targeted therapy of cancer.
Collapse
Affiliation(s)
- Bowei Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
| | - Jianjun Dong
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
| | - Fan Yang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
| | - Tuoyu Ju
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
| | - Junxi Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
| | - Kaige Qu
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
| | - Ying Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
| | - Yanling Tian
- School of Engineering, University of Warwick, Coventry CV4 7AL, UK
| | - Zuobin Wang
- International Research Centre for Nano Handling and Manufacturing of China, Changchun University of Science and Technology, Changchun 130022, China.
- Centre for Opto/Bio-Nano Measurement and Manufacturing, Zhongshan Institute of Changchun University of Science and Technology, Zhongshan 528437, China
- JR3CN & IRAC, University of Bedfordshire, Luton LU1 3JU, UK
| |
Collapse
|
9
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
10
|
Chen X, Xu Z, Tang K, Hu G, Du P, Wang J, Zhang C, Xin Y, Li K, Zhang Q, Hu J, Zhang Z, Yang M, Wang G, Tan Y. The Mechanics of Tumor Cells Dictate Malignancy via Cytoskeleton-Mediated APC/Wnt/β-Catenin Signaling. RESEARCH (WASHINGTON, D.C.) 2023; 6:0224. [PMID: 37746658 PMCID: PMC10513157 DOI: 10.34133/research.0224] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/20/2023] [Indexed: 09/26/2023]
Abstract
Tumor cells progressively remodel cytoskeletal structures and reduce cellular stiffness during tumor progression, implicating the correlation between cell mechanics and malignancy. However, the roles of tumor cell cytoskeleton and the mechanics in tumor progression remain incompletely understood. We report that softening/stiffening tumor cells by targeting actomyosin promotes/suppresses self-renewal in vitro and tumorigenic potential in vivo. Weakening/strengthening actin cytoskeleton impairs/reinforces the interaction between adenomatous polyposis coli (APC) and β-catenin, which facilitates β-catenin nuclear/cytoplasmic localization. Nuclear β-catenin binds to the promoter of Oct4, which enhances its transcription that is crucial in sustaining self-renewal and malignancy. These results demonstrate that the mechanics of tumor cells dictate self-renewal through cytoskeleton-APC-Wnt/β-catenin-Oct4 signaling, which are correlated with tumor differentiation and patient survival. This study unveils an uncovered regulatory role of cell mechanics in self-renewal and malignancy, and identifies tumor cell mechanics as a hallmark not only for cancer diagnosis but also for mechanotargeting.
Collapse
Affiliation(s)
- Xi Chen
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Research Institute of Smart Ageing,
The Hong Kong Polytechnic University, Hong Kong, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Zichen Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants,
Bioengineering College of Chongqing University, Chongqing, 400030, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Kai Tang
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Guanshuo Hu
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Research Institute of Smart Ageing,
The Hong Kong Polytechnic University, Hong Kong, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Pengyu Du
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Junfang Wang
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Cunyu Zhang
- Research Institute of Smart Ageing,
The Hong Kong Polytechnic University, Hong Kong, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Ying Xin
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Keming Li
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Qiantang Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants,
Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Jianjun Hu
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Zhuxue Zhang
- Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, Guizhou, 550002, China
| | - Mo Yang
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants,
Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, 518057, China
- Research Institute of Smart Ageing,
The Hong Kong Polytechnic University, Hong Kong, China
- Department of Biomedical Engineering,
The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
11
|
López-Guajardo A, Zafar A, Al Hennawi K, Rossi V, Alrwaili A, Medcalf JD, Dunning M, Nordgren N, Pettersson T, Estabrook ID, Hawkins RJ, Gad AKB. Regulation of cellular contractile force, shape and migration of fibroblasts by oncogenes and Histone deacetylase 6. Front Mol Biosci 2023; 10:1197814. [PMID: 37564130 PMCID: PMC10411354 DOI: 10.3389/fmolb.2023.1197814] [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: 03/31/2023] [Accepted: 07/05/2023] [Indexed: 08/12/2023] Open
Abstract
The capacity of cells to adhere to, exert forces upon and migrate through their surrounding environment governs tissue regeneration and cancer metastasis. The role of the physical contractile forces that cells exert in this process, and the underlying molecular mechanisms are not fully understood. We, therefore, aimed to clarify if the extracellular forces that cells exert on their environment and/or the intracellular forces that deform the cell nucleus, and the link between these forces, are defective in transformed and invasive fibroblasts, and to indicate the underlying molecular mechanism of control. Confocal, Epifluorescence and Traction force microscopy, followed by computational analysis, showed an increased maximum contractile force that cells apply on their environment and a decreased intracellular force on the cell nucleus in the invasive fibroblasts, as compared to normal control cells. Loss of HDAC6 activity by tubacin-treatment and siRNA-mediated HDAC6 knockdown also reversed the reduced size and more circular shape and defective migration of the transformed and invasive cells to normal. However, only tubacin-mediated, and not siRNA knockdown reversed the increased force of the invasive cells on their surrounding environment to normal, with no effects on nuclear forces. We observed that the forces on the environment and the nucleus were weakly positively correlated, with the exception of HDAC6 siRNA-treated cells, in which the correlation was weakly negative. The transformed and invasive fibroblasts showed an increased number and smaller cell-matrix adhesions than control, and neither tubacin-treatment, nor HDAC6 knockdown reversed this phenotype to normal, but instead increased it further. This highlights the possibility that the control of contractile force requires separate functions of HDAC6, than the control of cell adhesions, spreading and shape. These data are consistent with the possibility that defective force-transduction from the extracellular environment to the nucleus contributes to metastasis, via a mechanism that depends upon HDAC6. To our knowledge, our findings present the first correlation between the cellular forces that deforms the surrounding environment and the nucleus in fibroblasts, and it expands our understanding of how cells generate contractile forces that contribute to cell invasion and metastasis.
Collapse
Affiliation(s)
- Ana López-Guajardo
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Azeer Zafar
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Khairat Al Hennawi
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Valentina Rossi
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Abdulaziz Alrwaili
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Jessica D. Medcalf
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Mark Dunning
- Bioinformatics Core, The Medical School, The University of Sheffield, Sheffield, United Kingdom
| | - Niklas Nordgren
- Division Bioeconomy and Health, RISE Research Institutes of Sweden, Stockholm, Sweden
| | - Torbjörn Pettersson
- Fibre and Polymer Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Ian D. Estabrook
- Department of Physics and Astronomy, University of Sheffield, Sheffield, United Kingdom
- Center for Advancing Electronics Dresden, Technische Universität Dresden, Dresden, Germany
| | - Rhoda J. Hawkins
- Department of Physics and Astronomy, University of Sheffield, Sheffield, United Kingdom
- African Institute for Mathematical Sciences, Accra, Ghana
| | - Annica K. B. Gad
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
- Madeira Chemistry Research Centre, University of Madeira, Funchal, Portugal
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Millán M, Villarreal L, D'Aiuto N, Bologna-Molina R, Sotelo-Silveira J, Benech JC, Hochmann J, Arocena M. Mechanical profile of human keratinocytes expressing HPV-18 oncogenes. Biochem Biophys Res Commun 2023; 657:86-91. [PMID: 36996545 DOI: 10.1016/j.bbrc.2023.03.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
During tumorigenesis, the mechanical properties of cancer cells change markedly, with decreased stiffness often accompanying a more invasive phenotype. Less is known about the changes in mechanical parameters at intermediate stages in the process of malignant transformation. We have recently developed a pre-tumoral cell model by stably transducing the immortalized but non-tumorigenic human keratinocyte cell line HaCaT with the E5, E6 and E7 oncogenes from HPV-18, one of the leading causes of cervical cancer and other types of cancer worldwide. We have used atomic force microscopy (AFM) to measure cell stiffness and to obtain mechanical maps of parental HaCaT and HaCaT E5/E6/E7-18 cell lines. We observed a significant decrease in Young's modulus in HaCaT E5/E6/E7-18 cells measured by nanoindentation in the central region, as well as decreased cell rigidity in regions of cell-cell contact measured by Peakforce Quantitative Nanomechanical Mapping (PF-QNM). As a morphological correlate, HaCaT E5/E6/E7-18 cells displayed a significantly rounder cell shape than parental HaCaT cells. Our results therefore show that decreased stiffness with concomitant perturbations in cell shape are early mechanical and morphological changes during the process of malignant transformation.
Collapse
Affiliation(s)
- Magdalena Millán
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay
| | - Lihuén Villarreal
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay
| | - Natali D'Aiuto
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Cátedra de Bioquímica y Biofísica, Facultad de Odontología, Universidad de la República, Uruguay
| | - Ronell Bologna-Molina
- Departamento de Patología Molecular, Facultad de Odontología, Universidad de la República, Uruguay
| | - José Sotelo-Silveira
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Sección Biología Celular, Facultad de Ciencias, Universidad de la República, Uruguay
| | - Juan C Benech
- Laboratorio de Señalización Celular y Nanobiología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Plataforma de Microscopía de Fuerza Atómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay
| | - Jimena Hochmann
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Uruguay.
| | - Miguel Arocena
- Departamento de Genómica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Uruguay; Cátedra de Bioquímica y Biofísica, Facultad de Odontología, Universidad de la República, Uruguay.
| |
Collapse
|
13
|
Aguilar VM, Paul A, Lazarko D, Levitan I. Paradigms of endothelial stiffening in cardiovascular disease and vascular aging. Front Physiol 2023; 13:1081119. [PMID: 36714307 PMCID: PMC9874005 DOI: 10.3389/fphys.2022.1081119] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Endothelial cells, the inner lining of the blood vessels, are well-known to play a critical role in vascular function, while endothelial dysfunction due to different cardiovascular risk factors or accumulation of disruptive mechanisms that arise with aging lead to cardiovascular disease. In this review, we focus on endothelial stiffness, a fundamental biomechanical property that reflects cell resistance to deformation. In the first part of the review, we describe the mechanisms that determine endothelial stiffness, including RhoA-dependent contractile response, actin architecture and crosslinking, as well as the contributions of the intermediate filaments, vimentin and lamin. Then, we review the factors that induce endothelial stiffening, with the emphasis on mechanical signals, such as fluid shear stress, stretch and stiffness of the extracellular matrix, which are well-known to control endothelial biomechanics. We also describe in detail the contribution of lipid factors, particularly oxidized lipids, that were also shown to be crucial in regulation of endothelial stiffness. Furthermore, we discuss the relative contributions of these two mechanisms of endothelial stiffening in vasculature in cardiovascular disease and aging. Finally, we present the current state of knowledge about the role of endothelial stiffening in the disruption of endothelial cell-cell junctions that are responsible for the maintenance of the endothelial barrier.
Collapse
Affiliation(s)
- Victor M. Aguilar
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Amit Paul
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Dana Lazarko
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary and Critical Care, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
14
|
Kim HR, Warrington SJ, López-Guajardo A, Al Hennawi K, Cook SL, Griffith ZDJ, Symmes D, Zhang T, Qu Z, Xu Y, Chen R, Gad AKB. ALD-R491 regulates vimentin filament stability and solubility, cell contractile force, cell migration speed and directionality. Front Cell Dev Biol 2022; 10:926283. [PMID: 36483676 PMCID: PMC9723350 DOI: 10.3389/fcell.2022.926283] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 11/07/2022] [Indexed: 08/12/2023] Open
Abstract
Metastasizing cells express the intermediate filament protein vimentin, which is used to diagnose invasive tumors in the clinic. However, the role of vimentin in cell motility, and if the assembly of non-filamentous variants of vimentin into filaments regulates cell migration remains unclear. We observed that the vimentin-targeting drug ALD-R491 increased the stability of vimentin filaments, by reducing filament assembly and/or disassembly. ALD-R491-treatment also resulted in more bundled and disorganized filaments and an increased pool of non-filamentous vimentin. This was accompanied by a reduction in size of cell-matrix adhesions and increased cellular contractile forces. Moreover, during cell migration, cells showed erratic formation of lamellipodia at the cell periphery, loss of coordinated cell movement, reduced cell migration speed, directionality and an elongated cell shape with long thin extensions at the rear that often detached. Taken together, these results indicate that the stability of vimentin filaments and the soluble pool of vimentin regulate the speed and directionality of cell migration and the capacity of cells to migrate in a mechanically cohesive manner. These observations suggest that the stability of vimentin filaments governs the adhesive, physical and migratory properties of cells, and expands our understanding of vimentin functions in health and disease, including cancer metastasis.
Collapse
Affiliation(s)
- Hyejeong Rosemary Kim
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | | | - Ana López-Guajardo
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Khairat Al Hennawi
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Sarah L. Cook
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Zak D. J. Griffith
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Deebie Symmes
- Aluda Pharmaceuticals, Inc., Menlo Park, CA, United States
| | - Tao Zhang
- Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, China
| | - Zhipeng Qu
- Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, China
| | - Ying Xu
- Cambridge-Su Genomic Resource Center, Medical School of Soochow University, Suzhou, China
| | - Ruihuan Chen
- Aluda Pharmaceuticals, Inc., Menlo Park, CA, United States
| | - Annica K. B. Gad
- Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom
- Madeira Chemistry Research Centre, University of Madeira, Funchal, Portugal
| |
Collapse
|
15
|
Wang Z, Wang S, Jia Z, Zhao Y, Yang M, Yan W, Chen T, Xiang D, Shao R, Liu Y. Establishment and characterization of an immortalized epithelial cell line from human gallbladder. Front Oncol 2022; 12:994087. [PMID: 36387215 PMCID: PMC9650220 DOI: 10.3389/fonc.2022.994087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/26/2022] [Indexed: 11/25/2022] Open
Abstract
Background Although a plethora of studies have employed multiple gallbladder cancer (GBC) cell lines, it is surprisingly noted that there is still lack of a normal gallbladder epithelial cell line as a normal counterpart, thus impeding substantially the progress of mechanistic studies on the transformation of normal epithelial cells to cancer. Here, we created a normal gallbladder epithelial cell line named L-2F7 from human gallbladder tissue. Methods Gallbladder tissues from a diagnosed cholecystitis female patient were collected, and epithelial cells were enriched by magnetic cell sorting. Then, the cells were immortalized by co-introduction of human telomerase reverse transcriptase (hTERT) and Simian virus 40 large T antigen (LT-SV40) via a lentivirus infection system. After clonal selection and isolation, L-2F7 cells were tested for epithelial markers CK7, CK19, CK20, and CD326, genomic feature, cell proliferation, and migration using Western blot, immunofluorescence, whole genome sequencing, karyotyping, and RNA sequencing. L-2F7 cells were also transplanted to Nude (nu/nu) mice to determine tumorigenicity. Results We successfully identified one single-cell clone named L-2F7 which highly expressed epithelial markers CD326, CK7, CK19, and CK20. This cell line proliferated with a doubling time of 23 h and the epithelial morphology sustained over 30 passages following immortalization. Transient gene transduction of L-2F7 cells led to expression of exogenous GFP and FLAG protein. L-2F7 cells exhibited both distinct non-synonymous mutations from those of gallbladder cancer tissues and differential non-cancerous gene expression patterns similar to normal tissue. Although they displayed unexpected mobility, L-2F7 cells still lacked the ability to develop tumors. Conclusion We developed a non-cancerous gallbladder epithelial cell line, offering a valuable system for the study of gallbladder cancer and other gallbladder-related disorders.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
| | - Shijia Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
| | - Ziheng Jia
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
| | - Yuhao Zhao
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
| | - Mao Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
| | - Weikang Yan
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
| | - Tao Chen
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
| | - Dongxi Xiang
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
- *Correspondence: Dongxi Xiang, ; Rong Shao, ; Yingbin Liu,
| | - Rong Shao
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Dongxi Xiang, ; Rong Shao, ; Yingbin Liu,
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
- Shanghai Key Laboratory of Biliary Tract Disease, Renji Hospital, Shanghai, China
- Shanghai Research Center of Biliary Tract Disease, Renji Hospital, Shanghai, China
- *Correspondence: Dongxi Xiang, ; Rong Shao, ; Yingbin Liu,
| |
Collapse
|
16
|
How do cells stiffen? Biochem J 2022; 479:1825-1842. [PMID: 36094371 DOI: 10.1042/bcj20210806] [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: 05/11/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022]
Abstract
Cell stiffness is an important characteristic of cells and their response to external stimuli. In this review, we survey methods used to measure cell stiffness, summarize stimuli that alter cell stiffness, and discuss signaling pathways and mechanisms that control cell stiffness. Several pathological states are characterized by changes in cell stiffness, suggesting this property can serve as a potential diagnostic marker or therapeutic target. Therefore, we consider the effect of cell stiffness on signaling and growth processes required for homeostasis and dysfunction in healthy and pathological states. Specifically, the composition and structure of the cell membrane and cytoskeleton are major determinants of cell stiffness, and studies have identified signaling pathways that affect cytoskeletal dynamics both directly and by altered gene expression. We present the results of studies interrogating the effects of biophysical and biochemical stimuli on the cytoskeleton and other cellular components and how these factors determine the stiffness of both individual cells and multicellular structures. Overall, these studies represent an intersection of the fields of polymer physics, protein biochemistry, and mechanics, and identify specific mechanisms involved in mediating cell stiffness that can serve as therapeutic targets.
Collapse
|
17
|
Firnau MB, Brieger A. CK2 and the Hallmarks of Cancer. Biomedicines 2022; 10:1987. [PMID: 36009534 PMCID: PMC9405757 DOI: 10.3390/biomedicines10081987] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer is a leading cause of death worldwide. Casein kinase 2 (CK2) is commonly dysregulated in cancer, impacting diverse molecular pathways. CK2 is a highly conserved serine/threonine kinase, constitutively active and ubiquitously expressed in eukaryotes. With over 500 known substrates and being estimated to be responsible for up to 10% of the human phosphoproteome, it is of significant importance. A broad spectrum of diverse types of cancer cells has been already shown to rely on disturbed CK2 levels for their survival. The hallmarks of cancer provide a rationale for understanding cancer's common traits. They constitute the maintenance of proliferative signaling, evasion of growth suppressors, resisting cell death, enabling of replicative immortality, induction of angiogenesis, the activation of invasion and metastasis, as well as avoidance of immune destruction and dysregulation of cellular energetics. In this work, we have compiled evidence from the literature suggesting that CK2 modulates all hallmarks of cancer, thereby promoting oncogenesis and operating as a cancer driver by creating a cellular environment favorable to neoplasia.
Collapse
Affiliation(s)
| | - Angela Brieger
- Department of Internal Medicine I, Biomedical Research Laboratory, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| |
Collapse
|
18
|
Biomechanics of cancer stem cells. Essays Biochem 2022; 66:359-369. [PMID: 35942932 DOI: 10.1042/ebc20220014] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 12/27/2022]
Abstract
Cancer stem cells (CSCs) have been believed to be one driving force for tumor progression and drug resistance. Despite the significance of biochemical signaling in malignancy, highly malignant tumor cells or CSCs exhibit lower cellular stiffness than weakly malignant cells or non-CSCs, which are softer than their healthy counterparts, suggesting the inverse correlation between cell stiffness and malignancy. Recent years have witnessed the rapid accumulation of evidence illustrating the reciprocity between cell cytoskeleton/mechanics and CSC functions and the potential of cellular stiffness for specific targeting of CSCs. However, a systematic understanding of tumor cell mechanics and their role in CSCs and tumor progression is still lacking. The present review summarizes the recent progress in the alterations of tumor cell cytoskeleton and stiffness at different stages of tumor progression and recapitulates the relationship between cellular stiffness and CSC functions. The altered cell mechanics may mediate the mechanoadaptive responses that possibly empower CSCs to survive and thrive during metastasis. Furthermore, we highlight the possible impact of tumor cell mechanics on CSC malignancy, which may potentiate low cell stiffness as a mechanical marker for CSC targeting.
Collapse
|
19
|
Infante E, Etienne-Manneville S. Intermediate filaments: Integration of cell mechanical properties during migration. Front Cell Dev Biol 2022; 10:951816. [PMID: 35990612 PMCID: PMC9389290 DOI: 10.3389/fcell.2022.951816] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/07/2022] [Indexed: 11/22/2022] Open
Abstract
Cell migration is a vital and dynamic process required for the development of multicellular organisms and for immune system responses, tissue renewal and wound healing in adults. It also contributes to a variety of human diseases such as cancers, autoimmune diseases, chronic inflammation and fibrosis. The cytoskeleton, which includes actin microfilaments, microtubules, and intermediate filaments (IFs), is responsible for the maintenance of animal cell shape and structural integrity. Each cytoskeletal network contributes its unique properties to dynamic cell behaviour, such as cell polarization, membrane protrusion, cell adhesion and contraction. Hence, cell migration requires the dynamic orchestration of all cytoskeleton components. Among these, IFs have emerged as a molecular scaffold with unique mechanical features and a key player in the cell resilience to mechanical stresses during migration through complex 3D environment. Moreover, accumulating evidence illustrates the participation of IFs in signalling cascades and cytoskeletal crosstalk. Teaming up with actin and microtubules, IFs contribute to the active generation of forces required for cell adhesion and mesenchymal migration and invasion. Here we summarize and discuss how IFs integrate mechanical properties and signalling functions to control cell migration in a wide spectrum of physiological and pathological situations.
Collapse
Affiliation(s)
- Elvira Infante
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Université Paris-Cité, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Sandrine Etienne-Manneville
- Cell Polarity, Migration and Cancer Unit, Institut Pasteur, UMR3691 CNRS, Université Paris-Cité, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| |
Collapse
|
20
|
Abstract
More than 27 yr ago, the vimentin knockout (Vim-/- ) mouse was reported to develop and reproduce without an obvious phenotype, implying that this major cytoskeletal protein was nonessential. Subsequently, comprehensive and careful analyses have revealed numerous phenotypes in Vim-/- mice and their organs, tissues, and cells, frequently reflecting altered responses in the recovery of tissues following various insults or injuries. These findings have been supported by cell-based experiments demonstrating that vimentin intermediate filaments (IFs) play a critical role in regulating cell mechanics and are required to coordinate mechanosensing, transduction, signaling pathways, motility, and inflammatory responses. This review highlights the essential functions of vimentin IFs revealed from studies of Vim-/- mice and cells derived from them.
Collapse
Affiliation(s)
- Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois 60611, USA
- Department of Cell and Developmental Biology, Northwestern University, Chicago, Illinois 60611, USA
| | - John E Eriksson
- Cell Biology, Faculty of Science and Technology, Åbo Akademi University, FIN-20521 Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FIN-20521 Turku, Finland
- Euro-Bioimaging European Research Infrastructure Consortium (ERIC), FIN-20521 Turku, Finland
| | - Milos Pekny
- Laboratory of Astrocyte Biology and CNS Regeneration, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, 413 90 Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, Victoria 3052, Australia
- University of Newcastle, Newcastle, New South Wales 2300, Australia
| | - Robert D Goldman
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, Illinois 60611, USA
- Department of Cell and Developmental Biology, Northwestern University, Chicago, Illinois 60611, USA
| |
Collapse
|
21
|
Morris J, Takahashi-Ruiz L, Persi LN, Summers JC, McCauley EP, Chan PYW, Amberchan G, Lizama-Chamu I, Coppage DA, Crews P, Risinger AL, Johnson TA. Re-evaluation of the Fijianolide/Laulimalide Chemotype Suggests an Alternate Mechanism of Action for C-15/C-20 Analogs. ACS OMEGA 2022; 7:8824-8832. [PMID: 35309480 PMCID: PMC8928504 DOI: 10.1021/acsomega.1c07146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/23/2022] [Indexed: 06/14/2023]
Abstract
Herein, we report on naturally derived microtubule stabilizers with activity against triple negative breast cancer (TNBC) cell lines, including paclitaxel, fijianolide B/laulimalide (3), fijianolide B di-acetate (4), and two new semisynthetic analogs of 3, which include fijianolide J (5) and fijianolide L (6). Similar to paclitaxel, compound 3 demonstrated classic microtubule stabilizing activity with potent (GI50 = 0.7-17 nM) antiproliferative efficacy among the five molecularly distinct TNBC cell lines. Alternatively, compounds 5 or 6, generated from oxidation of C-20 or C-15 and C-20 respectively, resulted in a unique profile with reduced potency (GI50 = 4-9 μM), but improved efficacy in some lines, suggesting a distinct mechanism of action. The C-15, C-20 di-acetate, and dioxo modifications on 4 and 6 resulted in compounds devoid of classic microtubule stabilizing activity in biochemical assays. While 4 also had no detectable effect on cellular microtubules, 6 promoted a reorganization of the cytoskeleton resulting in an accumulation of microtubules at the cell periphery. Compound 5, with a single C-20 oxo substitution, displayed a mixed phenotype, sharing properties of 3 and 6. These results demonstrate the importance of the C-15/C-20 chiral centers, which appear to be required for the potent microtubule stabilizing activity of this chemotype and that oxidation of these sites promotes unanticipated cytoskeletal alterations that are distinct from classic microtubule stabilization, likely through a distinct mechanism of action.
Collapse
Affiliation(s)
- Joseph
D. Morris
- Department
of Natural Sciences, Dominican University
of California, San Rafael, California 94901, United States
| | - Leila Takahashi-Ruiz
- Department
of Pharmacology, University of Texas Health
Science Center at San Antonio, San Antonio, Texas 78229, United States
| | - Lauren N. Persi
- Department
of Natural Sciences, Dominican University
of California, San Rafael, California 94901, United States
| | - Jonathan C. Summers
- Department
of Natural Sciences, Dominican University
of California, San Rafael, California 94901, United States
| | - Erin P. McCauley
- Department
of Chemistry & Biochemistry, University
of California, Santa Cruz, California 95064, United States
| | - Peter Y. W. Chan
- Department
of Pharmacology, University of Texas Health
Science Center at San Antonio, San Antonio, Texas 78229, United States
| | - Gabriella Amberchan
- Department
of Chemistry & Biochemistry, University
of California, Santa Cruz, California 95064, United States
| | - Itzel Lizama-Chamu
- Department
of Chemistry & Biochemistry, University
of California, Santa Cruz, California 95064, United States
| | - David A. Coppage
- Department
of Chemistry & Biochemistry, University
of California, Santa Cruz, California 95064, United States
| | - Phillip Crews
- Department
of Chemistry & Biochemistry, University
of California, Santa Cruz, California 95064, United States
| | - April L. Risinger
- Department
of Pharmacology, University of Texas Health
Science Center at San Antonio, San Antonio, Texas 78229, United States
| | - Tyler A. Johnson
- Department
of Natural Sciences, Dominican University
of California, San Rafael, California 94901, United States
- Department
of Chemistry & Biochemistry, University
of California, Santa Cruz, California 95064, United States
| |
Collapse
|
22
|
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
|
23
|
Pai JT, Chen XH, Leu YL, Weng MS. Propolin G-Suppressed Epithelial-to-Mesenchymal Transition in Triple-Negative Breast Cancer Cells via Glycogen Synthase Kinase 3β-Mediated Snail and HDAC6-Regulated Vimentin Degradation. Int J Mol Sci 2022; 23:ijms23031672. [PMID: 35163593 PMCID: PMC8835855 DOI: 10.3390/ijms23031672] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 01/08/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer with a poor prognosis. The incidence and mortality rate of TNBC are frequently found in younger women. Due to the absence of a good therapeutic strategy, effective remedies for inhibiting TNBC have been developed for improving the cure rate. Epithelial-to-mesenchymal transition (EMT) is a critical mechanism to regulate cancer cell motility and invasion. Furthermore, ectopic expression of EMT molecules correlates with the metastasis and poor prognosis of TNBC. Targeting EMT might be a strategy for the therapy and prevention of TNBC. Propolin G, an active c-prenylflavanone in Taiwanese propolis, has been shown to possess anti-cancer activity in many cancers. However, the anti-metastasis activity of propolin G on TNBC is still unclear. The present study showed that the migration and invasion activities of TNBC cells was suppressed by propolin G. Down-regulated expression of Snail and vimentin and up-regulated expression of E-cadherin were dose- and time-dependently observed in propolin G-treated MDA-MB-231 cells. Propolin G inhibited Snail and vimentin expressions via the signaling pathways associated with post-translational modification. The activation of glycogen synthase kinase 3β (GSK-3β) by propolin G resulted in increasing GSK-3β interaction with Snail. Consequently, the nuclear localization and stability of Snail was disrupted resulting in promoting the degradation. Propolin G-inhibited Snail expression and the activities of migration and invasion were reversed by GSK-3β inhibitor pretreatment. Meanwhile, the outcomes also revealed that histone deacetylase 6 (HDAC6) activity was dose-dependently suppressed by propolin G. Correspondently, the amounts of acetyl-α-tubulin, a down-stream substrate of HDAC6, were increased. Dissociation of HDAC6/Hsp90 with vimentin leading to increased vimentin acetylation and degradation was perceived in the cells with the addition of propolin G. Moreover, up-regulated expression of acetyl-α-tubulin by propolin G was attenuated by HDAC6 overexpression. On the contrary, down-regulated expression of vimentin, cell migration and invasion by propolin G were overturned by HDAC6 overexpression. Conclusively, restraint cell migration and invasion of TNBC by propolin G were activated by the expression of GSK-3β-suppressed Snail and the interruption of HDAC6-mediated vimentin protein stability. Aiming at EMT, propolin G might be a potential candidate for TNBC therapy.
Collapse
Affiliation(s)
- Jih-Tung Pai
- Division of Hematology and Oncology, Tao-Yuan General Hospital, Ministry of Health and Welfare, Taoyuan City 33004, Taiwan;
| | - Xing-Han Chen
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
| | - Yann-Lii Leu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan;
- Tissue Bank, Chang Gung Memorial Hospital, Linkou, Taoyuan City 33342, Taiwan
| | - Meng-Shih Weng
- Department of Nutritional Science, Fu Jen Catholic University, New Taipei City 24205, Taiwan;
- Correspondence: ; Tel.: +886-2-2905-3776; Fax: +886-2-2902-1215
| |
Collapse
|
24
|
Zheng X, Li Z, Li W, Zhu M, Zhang L, Zhu Z, Yang H. Biomechanical properties of erythrocytes circulating in artificial hearts measured by dielectrophoretic method. J Biomech 2021; 129:110822. [PMID: 34736085 DOI: 10.1016/j.jbiomech.2021.110822] [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: 07/06/2021] [Revised: 09/28/2021] [Accepted: 10/16/2021] [Indexed: 11/16/2022]
Abstract
Blood damage is recognized as one of the major problems caused by non-physiological shear force induced by artificial hearts. At present, the generally accepted manifestation of mechanical blood damage is the amount of free hemoglobin released into the blood. However, there is little research on the changes of blood cell state after circulating in artificial hearts at the single-cell level. It is well known that the mechanical properties of cells are of enormous relevance in the regulation of cellular physiological and pathological processes. In this regard, it is highly needed to study the mechanical properties of blood cells affected by non-physiological shear force. In this paper, a dielectrophoresis-based method of measuring the mechanical properties of erythrocytes circulating in artificial hearts was proposed, which was quantified with some crucial parameters such as strain, elongation index (EI), and Young's modulus. Experimental results indicated that with the increase of the working time of artificial hearts, the deformability of erythrocytes decreased, the stiffness substantially increased, and the mechanical stability decreased, particularly at long exposure times. The proposed method provides a deep insight into the mechanism of subhemolytic damage at the single-cell level and has a great potential to serve as a new tool for in vitro evaluation of potential blood damage in artificial hearts.
Collapse
Affiliation(s)
- Xinyu Zheng
- Medical College of Soochow University, China
| | - Zhiwei Li
- Robotics and Microsystems Center, School of Mechanical and Electric Engineering, Soochow University, China
| | - Wanting Li
- Robotics and Microsystems Center, School of Mechanical and Electric Engineering, Soochow University, China
| | - Mingjie Zhu
- Robotics and Microsystems Center, School of Mechanical and Electric Engineering, Soochow University, China
| | - Liudi Zhang
- Artificial Organ Technology Lab, School of Mechanical and Electric Engineering, Soochow University, China
| | - Zhenhong Zhu
- Children's Hospital of Soochow University, China.
| | - Hao Yang
- Robotics and Microsystems Center, School of Mechanical and Electric Engineering, Soochow University, China.
| |
Collapse
|
25
|
Usman S, Waseem NH, Nguyen TKN, Mohsin S, Jamal A, Teh MT, Waseem A. Vimentin Is at the Heart of Epithelial Mesenchymal Transition (EMT) Mediated Metastasis. Cancers (Basel) 2021; 13:4985. [PMID: 34638469 PMCID: PMC8507690 DOI: 10.3390/cancers13194985] [Citation(s) in RCA: 229] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a reversible plethora of molecular events where epithelial cells gain the phenotype of mesenchymal cells to invade the surrounding tissues. EMT is a physiological event during embryogenesis (type I) but also happens during fibrosis (type II) and cancer metastasis (type III). It is a multifaceted phenomenon governed by the activation of genes associated with cell migration, extracellular matrix degradation, DNA repair, and angiogenesis. The cancer cells employ EMT to acquire the ability to migrate, resist therapeutic agents and escape immunity. One of the key biomarkers of EMT is vimentin, a type III intermediate filament that is normally expressed in mesenchymal cells but is upregulated during cancer metastasis. This review highlights the pivotal role of vimentin in the key events during EMT and explains its role as a downstream as well as an upstream regulator in this highly complex process. This review also highlights the areas that require further research in exploring the role of vimentin in EMT. As a cytoskeletal protein, vimentin filaments support mechanical integrity of the migratory machinery, generation of directional force, focal adhesion modulation and extracellular attachment. As a viscoelastic scaffold, it gives stress-bearing ability and flexible support to the cell and its organelles. However, during EMT it modulates genes for EMT inducers such as Snail, Slug, Twist and ZEB1/2, as well as the key epigenetic factors. In addition, it suppresses cellular differentiation and upregulates their pluripotent potential by inducing genes associated with self-renewability, thus increasing the stemness of cancer stem cells, facilitating the tumour spread and making them more resistant to treatments. Several missense and frameshift mutations reported in vimentin in human cancers may also contribute towards the metastatic spread. Therefore, we propose that vimentin should be a therapeutic target using molecular technologies that will curb cancer growth and spread with reduced mortality and morbidity.
Collapse
Affiliation(s)
- Saima Usman
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| | - Naushin H. Waseem
- UCL Institute of Ophthalmology, 11-43 Bath Str., London EC1V 9EL, UK;
| | - Thuan Khanh Ngoc Nguyen
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| | - Sahar Mohsin
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Ahmad Jamal
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| | - Muy-Teck Teh
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| | - Ahmad Waseem
- Centre for Oral Immunobiology and Regenerative Medicine, Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Turner Str., London E1 2AT, UK; (S.U.); (T.K.N.N.); (A.J.); (M.-T.T.)
| |
Collapse
|
26
|
Shi ML, Chen YF, Wu WQ, Lai Y, Jin Q, Qiu WL, Yu DL, Li YZ, Liao HF. Luteolin inhibits the proliferation, adhesion, migration and invasion of choroidal melanoma cells in vitro. Exp Eye Res 2021; 210:108643. [PMID: 34058231 DOI: 10.1016/j.exer.2021.108643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/24/2021] [Accepted: 05/24/2021] [Indexed: 12/27/2022]
Abstract
Choroidal melanoma is a devastating disease that causes visual loss and a high mortality rate due to metastasis. Luteolin, a potential anticancer compound, is widely found in natural plants. The aim of this study was to evaluate the antiproliferative, antiadhesive, antimigratory and anti-invasive effects of luteolin on choroidal melanoma cells in vitro and to explore its potential mechanism. Cell counting kit-8 (CCK-8) assays, 5-ethynyl-2'-deoxyuridine (EdU) assays, Cell adhesion, migration, and invasion assays were performed to examine the inhibitory effects of luteolin on cell cell viability, proliferation, adhesion, migration and invasion capacities, respectively. Considering the correlation between Matrix metalloenzymes and tumor metastasis, Enzyme-linked immunosorbent assays (ELISAs) were used to assess matrix metalloproteases MMP-2 and MMP-9 secretion. Western blotting was performed to detect p-PI3K P85, Akt, and p-Akt protein expression. The cytoskeletal proteins vimentin were observed with cell immunofluorescence staining. Luteolin can inhibit OCM-1 cell proliferation, migration, invasion and adhesion and C918 cell proliferation, migration, and invasion through the PI3K/Akt signaling pathway. Therefore, Luteolin may have potential as a therapeutic medication for Choroidal melanoma.
Collapse
Affiliation(s)
- Meng-Lin Shi
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Province Blood Center, Nanchang, 330052, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China
| | - Yu-Fen Chen
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Wei-Qi Wu
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Yao Lai
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Qi Jin
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Wan-Lu Qiu
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Dong-Lian Yu
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Yi-Zhong Li
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China
| | - Hong-Fei Liao
- Nanchang University, Nanchang, 330000, Jiangxi Province, China; Jiangxi Research Institute of Ophthalmology & Visual Sciences, Nanchang, 330006, Jiangxi Province, China; Department of Ophthalmology, The Affiliated Eye Hospital of Nanchang University, Nanchang, 330000, Jiangxi Province, China.
| |
Collapse
|
27
|
Runel G, Lopez-Ramirez N, Chlasta J, Masse I. Biomechanical Properties of Cancer Cells. Cells 2021; 10:cells10040887. [PMID: 33924659 PMCID: PMC8069788 DOI: 10.3390/cells10040887] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/24/2022] Open
Abstract
Since the crucial role of the microenvironment has been highlighted, many studies have been focused on the role of biomechanics in cancer cell growth and the invasion of the surrounding environment. Despite the search in recent years for molecular biomarkers to try to classify and stratify cancers, much effort needs to be made to take account of morphological and nanomechanical parameters that could provide supplementary information concerning tissue complexity adaptation during cancer development. The biomechanical properties of cancer cells and their surrounding extracellular matrix have actually been proposed as promising biomarkers for cancer diagnosis and prognosis. The present review first describes the main methods used to study the mechanical properties of cancer cells. Then, we address the nanomechanical description of cultured cancer cells and the crucial role of the cytoskeleton for biomechanics linked with cell morphology. Finally, we depict how studying interaction of tumor cells with their surrounding microenvironment is crucial to integrating biomechanical properties in our understanding of tumor growth and local invasion.
Collapse
Affiliation(s)
- Gaël Runel
- Centre de Recherche en Cancérologie de Lyon, CNRS-UMR5286, INSREM U1052, Université de Lyon, F-69008 Lyon, France; (G.R.); (N.L.-R.)
- BioMeca, F-69008 Lyon, France;
| | - Noémie Lopez-Ramirez
- Centre de Recherche en Cancérologie de Lyon, CNRS-UMR5286, INSREM U1052, Université de Lyon, F-69008 Lyon, France; (G.R.); (N.L.-R.)
| | | | - Ingrid Masse
- Centre de Recherche en Cancérologie de Lyon, CNRS-UMR5286, INSREM U1052, Université de Lyon, F-69008 Lyon, France; (G.R.); (N.L.-R.)
- Correspondence:
| |
Collapse
|
28
|
Baudoin NC, Bloomfield M. Karyotype Aberrations in Action: The Evolution of Cancer Genomes and the Tumor Microenvironment. Genes (Basel) 2021; 12:558. [PMID: 33921421 PMCID: PMC8068843 DOI: 10.3390/genes12040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/27/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is a disease of cellular evolution. For this cellular evolution to take place, a population of cells must contain functional heterogeneity and an assessment of this heterogeneity in the form of natural selection. Cancer cells from advanced malignancies are genomically and functionally very different compared to the healthy cells from which they evolved. Genomic alterations include aneuploidy (numerical and structural changes in chromosome content) and polyploidy (e.g., whole genome doubling), which can have considerable effects on cell physiology and phenotype. Likewise, conditions in the tumor microenvironment are spatially heterogeneous and vastly different than in healthy tissues, resulting in a number of environmental niches that play important roles in driving the evolution of tumor cells. While a number of studies have documented abnormal conditions of the tumor microenvironment and the cellular consequences of aneuploidy and polyploidy, a thorough overview of the interplay between karyotypically abnormal cells and the tissue and tumor microenvironments is not available. Here, we examine the evidence for how this interaction may unfold during tumor evolution. We describe a bidirectional interplay in which aneuploid and polyploid cells alter and shape the microenvironment in which they and their progeny reside; in turn, this microenvironment modulates the rate of genesis for new karyotype aberrations and selects for cells that are most fit under a given condition. We conclude by discussing the importance of this interaction for tumor evolution and the possibility of leveraging our understanding of this interplay for cancer therapy.
Collapse
Affiliation(s)
- Nicolaas C. Baudoin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Mathew Bloomfield
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
29
|
Graybill PM, Bollineni RK, Sheng Z, Davalos RV, Mirzaeifar R. A constriction channel analysis of astrocytoma stiffness and disease progression. BIOMICROFLUIDICS 2021; 15:024103. [PMID: 33763160 PMCID: PMC7968935 DOI: 10.1063/5.0040283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/23/2021] [Indexed: 05/12/2023]
Abstract
Studies have demonstrated that cancer cells tend to have reduced stiffness (Young's modulus) compared to their healthy counterparts. The mechanical properties of primary brain cancer cells, however, have remained largely unstudied. To investigate whether the stiffness of primary brain cancer cells decreases as malignancy increases, we used a microfluidic constriction channel device to deform healthy astrocytes and astrocytoma cells of grade II, III, and IV and measured the entry time, transit time, and elongation. Calculating cell stiffness directly from the experimental measurements is not possible. To overcome this challenge, finite element simulations of the cell entry into the constriction channel were used to train a neural network to calculate the stiffness of the analyzed cells based on their experimentally measured diameter, entry time, and elongation in the channel. Our study provides the first calculation of stiffness for grades II and III astrocytoma and is the first to apply a neural network analysis to determine cell mechanical properties from a constriction channel device. Our results suggest that the stiffness of astrocytoma cells is not well-correlated with the cell grade. Furthermore, while other non-central-nervous-system cell types typically show reduced stiffness of malignant cells, we found that most astrocytoma cell lines had increased stiffness compared to healthy astrocytes, with lower-grade astrocytoma having higher stiffness values than grade IV glioblastoma. Differences in nucleus-to-cytoplasm ratio only partly explain differences in stiffness values. Although our study does have limitations, our results do not show a strong correlation of stiffness with cell grade, suggesting that other factors may play important roles in determining the invasive capability of astrocytoma. Future studies are warranted to further elucidate the mechanical properties of astrocytoma across various pathological grades.
Collapse
Affiliation(s)
| | - R. K. Bollineni
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Z. Sheng
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine and Virginia Tech Fralin Biomedical Research Institute, Roanoke, Virginia 24016, USA
| | - R. V. Davalos
- Authors to whom correspondence should be addressed: and
| | - R. Mirzaeifar
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
30
|
Abstract
Vimentin is one of the first cytoplasmic intermediate filaments to be expressed in mammalian cells during embryogenesis, but its role in cellular fitness has long been a mystery. Vimentin is acknowledged to play a role in cell stiffness, cell motility, and cytoplasmic organization, yet it is widely considered to be dispensable for cellular function and organismal development. Here, we show that Vimentin plays a role in cellular stress response in differentiating cells, by recruiting aggregates, stress granules, and RNA-binding proteins, directing their elimination and asymmetric partitioning. In the absence of Vimentin, pluripotent embryonic stem cells fail to differentiate properly, with a pronounced deficiency in neuronal differentiation. Our results uncover a novel function for Vimentin, with important implications for development, tissue homeostasis, and in particular, stress response.
Collapse
|
31
|
LncRNA LINC00472 regulates cell stiffness and inhibits the migration and invasion of lung adenocarcinoma by binding to YBX1. Cell Death Dis 2020; 11:945. [PMID: 33144579 PMCID: PMC7609609 DOI: 10.1038/s41419-020-03147-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/18/2022]
Abstract
There is increasing evidence that long non-coding RNAs (lncRNAs) play important roles in human tumorigenesis. By using publicly available expression profiling data from lung adenocarcinoma and integrating bioinformatics analysis, we screened a lncRNA, LINC00472. LINC00472 expression in lung adenocarcinoma tissues was significantly lower and tightly associated with patient prognosis and TNM clinical stages in lung adenocarcinoma. LINC00472 also inhibited lung adenocarcinoma cell migration and invasion and increased cell stiffness and adhesion. RNA pull down and RIP assays identified that LINC00472 interacted with the transcription factor Y-box binding protein 1 (YBX1), which partially reversed the inhibition of cell migration and invasion and increased LINC00472-induced cell stiffness and adhesion. LINC00472 also regulated the density and integrity of F-actin in A549 and PC-9 cells possibly via YBX1. LINC00472 inhibited the cell epithelial-mesenchymal transition (EMT) processes via the modulation of YBX1. These results indicated that LINC00472 inhibited the cell EMT process by binding to YBX1, and affected the mechanical properties of the cell, ultimately inhibited its ability to invade and metastasize. Collectively, the present study provides the first evidence that LINC00472 changes the mechanical properties and inhibits the invasion and metastasis of lung adenocarcinoma cells.
Collapse
|
32
|
Seetharaman S, Etienne-Manneville S. Cytoskeletal Crosstalk in Cell Migration. Trends Cell Biol 2020; 30:720-735. [DOI: 10.1016/j.tcb.2020.06.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 01/15/2023]
|
33
|
The Major Capsid Protein, VP1, of the Mouse Polyomavirus Stimulates the Activity of Tubulin Acetyltransferase 1 by Microtubule Stabilization. Viruses 2020; 12:v12020227. [PMID: 32085463 PMCID: PMC7077302 DOI: 10.3390/v12020227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/17/2022] Open
Abstract
Viruses have evolved mechanisms to manipulate microtubules (MTs) for the efficient realization of their replication programs. Studying the mechanisms of replication of mouse polyomavirus (MPyV), we observed previously that in the late phase of infection, a considerable amount of the main structural protein, VP1, remains in the cytoplasm associated with hyperacetylated microtubules. VP1–microtubule interactions resulted in blocking the cell cycle in the G2/M phase. We are interested in the mechanism leading to microtubule hyperacetylation and stabilization and the roles of tubulin acetyltransferase 1 (αTAT1) and deacetylase histone deacetylase 6 (HDAC6) and VP1 in this mechanism. Therefore, HDAC6 inhibition assays, αTAT1 knock out cell infections, in situ cell fractionation, and confocal and TIRF microscopy were used. The experiments revealed that the direct interaction of isolated microtubules and VP1 results in MT stabilization and a restriction of their dynamics. VP1 leads to an increase in polymerized tubulin in cells, thus favoring αTAT1 activity. The acetylation status of MTs did not affect MPyV infection. However, the stabilization of MTs by VP1 in the late phase of infection may compensate for the previously described cytoskeleton destabilization by MPyV early gene products and is important for the observed inhibition of the G2→M transition of infected cells to prolong the S phase.
Collapse
|
34
|
Nguyen AV, Trompetto B, Tan XHM, Scott MB, Hu KHH, Deeds E, Butte MJ, Chiou PY, Rowat AC. Differential Contributions of Actin and Myosin to the Physical Phenotypes and Invasion of Pancreatic Cancer Cells. Cell Mol Bioeng 2020; 13:27-44. [PMID: 32030106 PMCID: PMC6981337 DOI: 10.1007/s12195-019-00603-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Metastasis is a fundamentally physical process in which cells deform through narrow gaps and generate forces to invade surrounding tissues. While it is commonly thought that increased cell deformability is an advantage for invading cells, we previously found that more invasive pancreatic ductal adenocarcinoma (PDAC) cells are stiffer than less invasive PDAC cells. Here we investigate potential mechanisms of the simultaneous increase in PDAC cell stiffness and invasion, focusing on the contributions of myosin II, Arp2/3, and formins. METHOD We measure cell invasion using a 3D scratch wound invasion assay and cell stiffness using atomic force microscopy (AFM). To determine the effects of actin- and myosin-mediated force generation on cell stiffness and invasion, we treat cells with pharmacologic inhibitors of myosin II (blebbistatin), Arp2/3 (CK-666), and formins (SMIFH2). RESULTS We find that the activity of myosin II, Arp2/3, and formins all contribute to the stiffness of PDAC cells. Interestingly, we find that the invasion of PDAC cell lines is differentially affected when the activity of myosin II, Arp2/3, or formins is inhibited, suggesting that despite having similar tissue origins, different PDAC cell lines may rely on different mechanisms for invasion. CONCLUSIONS These findings deepen our knowledge of the factors that regulate cancer cell mechanotype and invasion, and incite further studies to develop therapeutics that target multiple mechanisms of invasion for improved clinical benefit.
Collapse
Affiliation(s)
- Angelyn V. Nguyen
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
| | - Brittany Trompetto
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
| | | | - Michael B. Scott
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
- Present Address: Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, USA
- Department of Biomedical Engineering, Northwestern McCormick School of Engineering, Evanston, USA
| | | | - Eric Deeds
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, USA
| | - Manish J. Butte
- Department of Pediatrics, University of California, Los Angeles, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, USA
| | - Pei Yu Chiou
- Department of Bioengineering, University of California, Los Angeles, USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
| | - Amy C. Rowat
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, USA
| |
Collapse
|
35
|
Horníková L, Bruštíková K, Forstová J. Microtubules in Polyomavirus Infection. Viruses 2020; 12:E121. [PMID: 31963741 PMCID: PMC7019765 DOI: 10.3390/v12010121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/12/2022] Open
Abstract
Microtubules, part of the cytoskeleton, are indispensable for intracellular movement, cell division, and maintaining cell shape and polarity. In addition, microtubules play an important role in viral infection. In this review, we summarize the role of the microtubules' network during polyomavirus infection. Polyomaviruses usurp microtubules and their motors to travel via early and late acidic endosomes to the endoplasmic reticulum. As shown for SV40, kinesin-1 and microtubules are engaged in the release of partially disassembled virus from the endoplasmic reticulum to the cytosol, and dynein apparently assists in the further disassembly of virions prior to their translocation to the cell nucleus-the place of their replication. Polyomavirus gene products affect the regulation of microtubule dynamics. Early T antigens destabilize microtubules and cause aberrant mitosis. The role of these activities in tumorigenesis has been documented. However, its importance for productive infection remains elusive. On the other hand, in the late phase of infection, the major capsid protein, VP1, of the mouse polyomavirus, counteracts T-antigen-induced destabilization. It physically binds microtubules and stabilizes them. The interaction results in the G2/M block of the cell cycle and prolonged S phase, which is apparently required for successful completion of the viral replication cycle.
Collapse
Affiliation(s)
| | | | - Jitka Forstová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, 25250 Vestec, Czech Republic; (L.H.); (K.B.)
| |
Collapse
|
36
|
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
|
37
|
Kapsokalyvas D, van Zandvoort MAMJ. Molecular Imaging in Oncology: Advanced Microscopy Techniques. Recent Results Cancer Res 2020; 216:533-561. [PMID: 32594398 DOI: 10.1007/978-3-030-42618-7_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Preclinical studies usually require high levels of morphological, functional, and biochemical information at subcellular resolution. This type of information cannot be obtained from clinical imaging techniques, such as MRI, PET/CT, or US. Luckily, many microscopy techniques exist that can offer this information, also for malignant tissues and therapeutic approaches. In this overview, we discuss the various advanced optical microscopy techniques and their applications in oncological research. After a short introduction in Sect. 16.1, we continue in Sect. 16.2 with a discussion on fluorescent labelling strategies, followed in Sect. 16.3 by an in-depth description of confocal, light-sheet, two-photon, and super-resolution microscopy. We end in Sect. 16.4 with a focus on the applications, specifically in oncology.
Collapse
Affiliation(s)
- Dimitrios Kapsokalyvas
- School for Oncology and Developmental Biology GROW and School for Cardiovascular Diseases CARIM, Maastricht University, Maastricht, The Netherlands
- Institut für Molekulare Kreislaufforschung, Universitätsklinikum Aachen, Aachen, Germany
| | - Marc A M J van Zandvoort
- School for Oncology and Developmental Biology GROW and School for Cardiovascular Diseases CARIM, Maastricht University, Maastricht, The Netherlands.
- Institut für Molekulare Kreislaufforschung, Universitätsklinikum Aachen, Aachen, Germany.
| |
Collapse
|
38
|
Finite Element Modelling of Single Cell Based on Atomic Force Microscope Indentation Method. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2019; 2019:7895061. [PMID: 31933677 PMCID: PMC6942797 DOI: 10.1155/2019/7895061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/27/2019] [Indexed: 02/03/2023]
Abstract
The stiffness of cells, especially cancer cells, is a key mechanical property that is closely associated with their biomechanical functions, such as the mechanotransduction and the metastasis mechanisms of cancer cells. In light of the low survival rate of single cells and measurement uncertainty, the finite element method (FEM) was used to quantify the deformations and predict the stiffness of single cells. To study the effect of the cell components on overall stiffness, two new FEM models were proposed based on the atomic force microscopy (AFM) indentation method. The geometric sizes of the FEM models were determined by AFM topography images, and the validity of the FEM models was verified by comparison with experimental data. The effect of the intermediate filaments (IFs) and material properties of the cellular continuum components on the overall stiffness were investigated. The experimental results showed that the stiffness of cancer cells has apparent positional differences. The FEM simulation results show that IFs contribute only slightly to the overall stiffness within 10% strain, although they can transfer forces directly from the membrane to the nucleus. The cytoskeleton (CSK) is the major mechanical component of a cell. Furthermore, parameter studies revealed that the material properties (thickness and elasticity) of the continuum have a significant influence on the overall cellular stiffness while Poisson's ratio has less of an influence on the overall cellular stiffness. The proposed FEM models can determine the contribution of the major components of the cells to the overall cellular stiffness and provide insights for understanding the response of cells to the external mechanical stimuli and studying the corresponding mechanical mechanisms and cell biomechanics.
Collapse
|
39
|
Vohnoutka RB, Gulvady AC, Goreczny G, Alpha K, Handelman SK, Sexton JZ, Turner CE. The focal adhesion scaffold protein Hic-5 regulates vimentin organization in fibroblasts. Mol Biol Cell 2019; 30:3037-3056. [PMID: 31644368 PMCID: PMC6880880 DOI: 10.1091/mbc.e19-08-0442] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Focal adhesion (FA)-stimulated reorganization of the F-actin cytoskeleton regulates cellular size, shape, and mechanical properties. However, FA cross-talk with the intermediate filament cytoskeleton is poorly understood. Genetic ablation of the FA-associated scaffold protein Hic-5 in mouse cancer-associated fibroblasts (CAFs) promoted a dramatic collapse of the vimentin network, which was rescued following EGFP-Hic-5 expression. Vimentin collapse correlated with a loss of detergent-soluble vimentin filament precursors and decreased vimentin S72/S82 phosphorylation. Additionally, fluorescence recovery after photobleaching analysis indicated impaired vimentin dynamics. Microtubule (MT)-associated EB1 tracking and Western blotting of MT posttranslational modifications indicated no change in MT dynamics that could explain the vimentin collapse. However, pharmacological inhibition of the RhoGTPase Cdc42 in Hic-5 knockout CAFs rescued the vimentin collapse, while pan-formin inhibition with SMIFH2 promoted vimentin collapse in Hic-5 heterozygous CAFs. Our results reveal novel regulation of vimentin organization/dynamics by the FA scaffold protein Hic-5 via modulation of RhoGTPases and downstream formin activity.
Collapse
Affiliation(s)
- Rishel B Vohnoutka
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Anushree C Gulvady
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Gregory Goreczny
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Kyle Alpha
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Samuel K Handelman
- Division of Gastroenterology, Department of Internal Medicine, Michigan Medicine at the University of Michigan, Ann Arbor, MI 48109
| | - Jonathan Z Sexton
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
40
|
Udartseva OO, Zhidkova OV, Ezdakova MI, Ogneva IV, Andreeva ER, Buravkova LB, Gollnick SO. Low-dose photodynamic therapy promotes angiogenic potential and increases immunogenicity of human mesenchymal stromal cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 199:111596. [DOI: 10.1016/j.jphotobiol.2019.111596] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/23/2019] [Accepted: 08/14/2019] [Indexed: 12/19/2022]
|
41
|
Mónico A, Duarte S, Pajares MA, Pérez-Sala D. Vimentin disruption by lipoxidation and electrophiles: Role of the cysteine residue and filament dynamics. Redox Biol 2019; 23:101098. [PMID: 30658903 PMCID: PMC6859561 DOI: 10.1016/j.redox.2019.101098] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 12/28/2018] [Accepted: 01/05/2019] [Indexed: 12/17/2022] Open
Abstract
The intermediate filament protein vimentin constitutes a critical sensor for electrophilic and oxidative stress, which induce extensive reorganization of the vimentin cytoskeletal network. Here, we have investigated the mechanisms underlying these effects. In vitro, electrophilic lipids, including 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) and 4-hydroxynonenal (HNE), directly bind to vimentin, whereas the oxidant diamide induces disulfide bond formation. Mutation of the single vimentin cysteine residue (Cys328) blunts disulfide formation and reduces lipoxidation by 15d-PGJ2, but not HNE. Preincubation with these agents differentially hinders NaCl-induced filament formation by wild-type vimentin, with effects ranging from delayed elongation and increased filament diameter to severe impairment of assembly or aggregation. Conversely, the morphology of vimentin Cys328Ser filaments is mildly or not affected. Interestingly, preformed vimentin filaments are more resistant to electrophile-induced disruption, although chemical modification is not diminished, showing that vimentin (lip)oxidation prior to assembly is more deleterious. In cells, electrophiles, particularly diamide, induce a fast and drastic disruption of existing filaments, which requires the presence of Cys328. As the cellular vimentin network is under continuous remodeling, we hypothesized that vimentin exchange on filaments would be necessary for diamide-induced disruption. We confirmed that strategies reducing vimentin dynamics, as monitored by FRAP, including cysteine crosslinking and ATP synthesis inhibition, prevent diamide effect. In turn, phosphorylation may promote vimentin disassembly. Indeed, treatment with the phosphatase inhibitor calyculin A to prevent dephosphorylation intensifies electrophile-induced wild-type vimentin filament disruption. However, whereas a phosphorylation-deficient vimentin mutant is only partially protected from disorganization, Cys328Ser vimentin is virtually resistant, even in the presence of calyculin A. Together, these results indicate that modification of Cys328 and vimentin exchange are critical for electrophile-induced network disruption.
Collapse
Affiliation(s)
- Andreia Mónico
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Sofia Duarte
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María A Pajares
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain; Molecular Hepatology Group, Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain.
| |
Collapse
|
42
|
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
|
43
|
Nyberg KD, Bruce SL, Nguyen AV, Chan CK, Gill NK, Kim TH, Sloan EK, Rowat AC. Predicting cancer cell invasion by single-cell physical phenotyping. Integr Biol (Camb) 2019; 10:218-231. [PMID: 29589844 DOI: 10.1039/c7ib00222j] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The physical properties of cells are promising biomarkers for cancer diagnosis and prognosis. Here we determine the physical phenotypes that best distinguish human cancer cell lines, and their relationship to cell invasion. We use the high throughput, single-cell microfluidic method, quantitative deformability cytometry (q-DC), to measure six physical phenotypes including elastic modulus, cell fluidity, transit time, entry time, cell size, and maximum strain at rates of 102 cells per second. By training a k-nearest neighbor machine learning algorithm, we demonstrate that multiparameter analysis of physical phenotypes enhances the accuracy of classifying cancer cell lines compared to single parameters alone. We also discover a set of four physical phenotypes that predict invasion; using these four parameters, we generate the physical phenotype model of invasion by training a multiple linear regression model with experimental data from a set of human ovarian cancer cells that overexpress a panel of tumor suppressor microRNAs. We validate the model by predicting invasion based on measured physical phenotypes of breast and ovarian human cancer cell lines that are subject to genetic or pharmacologic perturbations. Taken together, our results highlight how physical phenotypes of single cells provide a biomarker to predict the invasion of cancer cells.
Collapse
Affiliation(s)
- Kendra D Nyberg
- Department of Integrative Biology and Physiology, University of California, 610 Charles E. Young Dr East, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Danielsson F, Peterson MK, Caldeira Araújo H, Lautenschläger F, Gad AKB. Vimentin Diversity in Health and Disease. Cells 2018; 7:E147. [PMID: 30248895 PMCID: PMC6210396 DOI: 10.3390/cells7100147] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/16/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022] Open
Abstract
Vimentin is a protein that has been linked to a large variety of pathophysiological conditions, including cataracts, Crohn's disease, rheumatoid arthritis, HIV and cancer. Vimentin has also been shown to regulate a wide spectrum of basic cellular functions. In cells, vimentin assembles into a network of filaments that spans the cytoplasm. It can also be found in smaller, non-filamentous forms that can localise both within cells and within the extracellular microenvironment. The vimentin structure can be altered by subunit exchange, cleavage into different sizes, re-annealing, post-translational modifications and interacting proteins. Together with the observation that different domains of vimentin might have evolved under different selection pressures that defined distinct biological functions for different parts of the protein, the many diverse variants of vimentin might be the cause of its functional diversity. A number of review articles have focussed on the biology and medical aspects of intermediate filament proteins without particular commitment to vimentin, and other reviews have focussed on intermediate filaments in an in vitro context. In contrast, the present review focusses almost exclusively on vimentin, and covers both ex vivo and in vivo data from tissue culture and from living organisms, including a summary of the many phenotypes of vimentin knockout animals. Our aim is to provide a comprehensive overview of the current understanding of the many diverse aspects of vimentin, from biochemical, mechanical, cellular, systems biology and medical perspectives.
Collapse
Affiliation(s)
- Frida Danielsson
- Science for Life Laboratory, Royal Institute of Technology, 17165 Stockholm, Sweden.
| | | | | | - Franziska Lautenschläger
- Campus D2 2, Leibniz-Institut für Neue Materialien gGmbH (INM) and Experimental Physics, NT Faculty, E 2 6, Saarland University, 66123 Saarbrücken, Germany.
| | - Annica Karin Britt Gad
- Centro de Química da Madeira, Universidade da Madeira, 9020105 Funchal, Portugal.
- Department of Medical Biochemistry and Microbiology, Uppsala University, 75237 Uppsala, Sweden.
| |
Collapse
|
45
|
Yumiyama S, Fujisawa E, Konishi Y, Nomura T. Control of colloidal behavior of polystyrene latex nanoparticles and their cytotoxicity toward yeast cells using water-soluble polymers. ADV POWDER TECHNOL 2018. [DOI: 10.1016/j.apt.2018.06.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
46
|
Li S, Xiong N, Peng Y, Tang K, Bai H, Lv X, Jiang Y, Qin X, Yang H, Wu C, Zhou P, Liu Y. Acidic pHe regulates cytoskeletal dynamics through conformational integrin β1 activation and promotes membrane protrusion. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2395-2408. [PMID: 29698684 DOI: 10.1016/j.bbadis.2018.04.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/02/2018] [Accepted: 04/19/2018] [Indexed: 12/15/2022]
Abstract
An acidic extracellular pH (pHe) in the tumor microenvironment has been suggested to facilitate tumor growth and metastasis. However, the molecular mechanisms by which tumor cells sense acidic signal to induce a transition to an aggressive phenotype remain elusive. Here, we showed that an acidic pHe (pH 6.5) stimulation resulted in protrusion and epithelial-mesenchymal transition (EMT) of cancer cells, which promoted migration and matrix degeneration. Using computational molecular dynamics simulations, we reported acidic pHe-induced opening of the Integrin dimers (α5β1) headpiece which indicated the activation of integrin. Moreover, acidic pHe promoted maturation of focal adhesions, temporal activation of Rho GTPases and microfilament reorganization through integrin β1-activated FAK signaling. Furthermore, mechanical balance of cytoskeleton (actin, tubulin and vimentin) contributed to acidic pHe-triggered protrusion and morphology change. Taken together, these findings revealed that integrin β1 could be a novel pH-regulated sensitive molecule which confers protrusion and malignant phenotype of cancer cells.
Collapse
Affiliation(s)
- Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Niya Xiong
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yueting Peng
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Kai Tang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hongxia Bai
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiaoying Lv
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Ying Jiang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Xiang Qin
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Peng Zhou
- Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China; Center for Information in Biology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, PR China.
| |
Collapse
|
47
|
Wang J, Liu M, Shen Y, Sun J, Shao Z, Czajkowsky DM. Compressive Force Spectroscopy: From Living Cells to Single Proteins. Int J Mol Sci 2018; 19:E960. [PMID: 29570665 PMCID: PMC5979447 DOI: 10.3390/ijms19040960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 12/20/2022] Open
Abstract
One of the most successful applications of atomic force microscopy (AFM) in biology involves monitoring the effect of force on single biological molecules, often referred to as force spectroscopy. Such studies generally entail the application of pulling forces of different magnitudes and velocities upon individual molecules to resolve individualistic unfolding/separation pathways and the quantification of the force-dependent rate constants. However, a less recognized variation of this method, the application of compressive force, actually pre-dates many of these "tensile" force spectroscopic studies. Further, beyond being limited to the study of single molecules, these compressive force spectroscopic investigations have spanned samples as large as living cells to smaller, multi-molecular complexes such as viruses down to single protein molecules. Correspondingly, these studies have enabled the detailed characterization of individual cell states, subtle differences between seemingly identical viral structures, as well as the quantification of rate constants of functionally important, structural transitions in single proteins. Here, we briefly review some of the recent achievements that have been obtained with compressive force spectroscopy using AFM and highlight exciting areas of its future development.
Collapse
Affiliation(s)
- Jiabin Wang
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Meijun Liu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yi Shen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jielin Sun
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Zhifeng Shao
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Daniel Mark Czajkowsky
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
48
|
Shukla VC, Kuang TR, Senthilvelan A, Higuita-Castro N, Duarte-Sanmiguel S, Ghadiali SN, Gallego-Perez D. Lab-on-a-Chip Platforms for Biophysical Studies of Cancer with Single-Cell Resolution. Trends Biotechnol 2018; 36:549-561. [PMID: 29559164 DOI: 10.1016/j.tibtech.2018.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/15/2018] [Accepted: 02/16/2018] [Indexed: 12/14/2022]
Abstract
Recent cancer research has more strongly emphasized the biophysical aspects of tumor development, progression, and microenvironment. In addition to genetic modifications and mutations in cancer cells, it is now well accepted that the physical properties of cancer cells such as stiffness, electrical impedance, and refractive index vary with tumor progression and can identify a malignant phenotype. Moreover, cancer heterogeneity renders population-based characterization techniques inadequate, as individual cellular features are lost in the average. Hence, platforms for fast and accurate characterization of biophysical properties of cancer cells at the single-cell level are required. Here, we highlight some of the recent advances in the field of cancer biophysics and the development of lab-on-a-chip platforms for single-cell biophysical analyses of cancer cells.
Collapse
Affiliation(s)
- Vasudha C Shukla
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine and Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; These authors contributed equally to this work
| | - Tai-Rong Kuang
- The Key Laboratory of Polymer Processing Engineering of Ministry of Education, South China University of Technology, Guangzhou 510640, P.R. China; These authors contributed equally to this work.
| | - Abirami Senthilvelan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Natalia Higuita-Castro
- Department of Internal Medicine (Division of Pulmonary, Critical Care and Sleep Medicine), Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Silvia Duarte-Sanmiguel
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Human Sciences (Human Nutrition), College of Human Ecology, The Ohio State University, Columbus, OH 43210, USA
| | - Samir N Ghadiali
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Internal Medicine (Division of Pulmonary, Critical Care and Sleep Medicine), Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH 43210, USA; Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
49
|
Rajagopal V, Holmes WR, Lee PVS. Computational modeling of single-cell mechanics and cytoskeletal mechanobiology. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2018; 10:e1407. [PMID: 29195023 PMCID: PMC5836888 DOI: 10.1002/wsbm.1407] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/19/2017] [Accepted: 09/07/2017] [Indexed: 01/10/2023]
Abstract
Cellular cytoskeletal mechanics plays a major role in many aspects of human health from organ development to wound healing, tissue homeostasis and cancer metastasis. We summarize the state-of-the-art techniques for mathematically modeling cellular stiffness and mechanics and the cytoskeletal components and factors that regulate them. We highlight key experiments that have assisted model parameterization and compare the advantages of different models that have been used to recapitulate these experiments. An overview of feed-forward mechanisms from signaling to cytoskeleton remodeling is provided, followed by a discussion of the rapidly growing niche of encapsulating feedback mechanisms from cytoskeletal and cell mechanics to signaling. We discuss broad areas of advancement that could accelerate research and understanding of cellular mechanobiology. A precise understanding of the molecular mechanisms that affect cell and tissue mechanics and function will underpin innovations in medical device technologies of the future. WIREs Syst Biol Med 2018, 10:e1407. doi: 10.1002/wsbm.1407 This article is categorized under: Models of Systems Properties and Processes > Mechanistic Models Physiology > Mammalian Physiology in Health and Disease Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Vijay Rajagopal
- Cell Structure and Mechanobiology Group, Department of Biomedical EngineeringUniversity of MelbourneMelbourneAustralia
| | - William R. Holmes
- Department of Physics and AstronomyVanderbilt UniversityNashvilleTNUSA
| | - Peter Vee Sin Lee
- Cell and Tissue Biomechanics Laboratory, Department of Biomedical EngineeringUniversity of MelbourneMelbourneAustralia
| |
Collapse
|
50
|
Long noncoding RNA LINC00675 enhances phosphorylation of vimentin on Ser83 to suppress gastric cancer progression. Cancer Lett 2018; 412:179-187. [DOI: 10.1016/j.canlet.2017.10.026] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 01/04/2023]
|