1
|
Swoger M, Ho Thanh MT, Patteson AE. Vimentin - Force regulator in confined environments. Curr Opin Cell Biol 2025; 94:102521. [PMID: 40288055 DOI: 10.1016/j.ceb.2025.102521] [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: 11/06/2024] [Revised: 03/16/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025]
Abstract
Cells must navigate crowded and confining 3D environments during normal function in vivo. Essential to their ability to navigate these environments safely and efficiently is their ability to mediate and endure both self-generated and external forces. The cytoskeleton, composed of F-actin, microtubules, and intermediate filaments, provides the mechanical support necessary for force mediation. The role of F-actin and microtubules in this process has been well studied, whereas vimentin, a cytoplasmic intermediate filament associated with mesenchymal cells, is less studied. However, there is growing evidence that vimentin has functions in both force transmission and protection of the cell from mechanical stress that actin and microtubules cannot fulfill. This review focuses on recent reports highlighting vimentin's role in regulating forces in confining environments.
Collapse
Affiliation(s)
- Maxx Swoger
- Department of Physics, Syracuse University, USA; BioInspired Institute, Syracuse University, USA; Department of Medicine, University of Pennsylvania, USA.
| | - Minh Tri Ho Thanh
- Department of Physics, Syracuse University, USA; BioInspired Institute, Syracuse University, USA
| | - Alison E Patteson
- Department of Physics, Syracuse University, USA; BioInspired Institute, Syracuse University, USA.
| |
Collapse
|
2
|
Lv CL, Li B. Interface morphodynamics in living tissues. SOFT MATTER 2025; 21:3670-3687. [PMID: 40226989 DOI: 10.1039/d5sm00145e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Interfaces between distinct tissues or between tissues and environments are common in multicellular organisms. The evolution and stability of these interfaces are essential for tissue development, and their dysfunction can lead to diseases such as cancer. Mounting efforts, either theoretical or experimental, have been devoted to uncovering the morphodynamics of tissue interfaces. Here, we review the recent progress of studies on interface morphodynamics. The regulatory mechanisms governing interface evolution are dissected, with a focus on adhesion, cortical tension, cell activity, extracellular matrix, and microenvironment. We examine the methodologies used to study morphodynamics, emphasizing the characteristics of experimental techniques and theoretical models. Finally, we explore the broader implications of interface morphodynamics in tissue morphogenesis and diseases, offering a comprehensive perspective on this rapidly developing field.
Collapse
Affiliation(s)
- Cheng-Lin Lv
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China.
| | - Bo Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China.
- Mechano-X Institute, Department of Engineering Mechanics, Tsinghua University, Beijing, China
- State Key Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing, China
| |
Collapse
|
3
|
Abdellatef SA, Wang H, Nakanishi J. Microtubules Disruption Alters the Cellular Structures and Mechanics Depending on Underlying Chemical Cues. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2312282. [PMID: 39344221 PMCID: PMC11962689 DOI: 10.1002/smll.202312282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 09/19/2024] [Indexed: 10/01/2024]
Abstract
The extracellular matrix determines cell morphology and stiffness by manipulating the cytoskeleton. The impacts of extracellular matrix cues, including the mechanical and topographical cues on microtubules and their role in biological behaviors, are previously studied. However, there is a lack of understanding about how microtubules (MTs) are affected by environmental chemical cues, such as extracellular matrix density. Specifically, it is crucial to understand the connection between cellular morphology and mechanics induced by chemical cues and the role of microtubules in these cellular responses. To address this, surfaces with high and low cRGD (cyclic Arginine-Glycine-Aspartic acid) peptide ligand densities are used. The cRGD is diluted with a bioinert ligand to prevent surface native cellular remodeling. The cellular morphology, actin, and microtubules differ on these surfaces. Confocal fluorescence microscopes and atomic force microscopy (AFM) are used to determine the structural and mechanical cellular responses with and without microtubules. Microtubules are vital as an intracellular scaffold in elongated morphology correlated with low cRGD compared to rounded morphology in high cRGD substrates. The contributions of MTs to nucleus morphology and cellular mechanics are based on the underlying cRGD densities. Finally, this study reveals a significant correlation between MTs, actin networks, and vimentin in response to the underlying densities of cRGD.
Collapse
Affiliation(s)
- Shimaa A. Abdellatef
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
| | - Hongxin Wang
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
| | - Jun Nakanishi
- Research Center for Macromolecules and BiomaterialsNational Institute for Materials Science (NIMS)1‐1 NamikiTsukuba305‐0044Japan
- Graduate School of Advanced EngineeringTokyo University of Science6‐3‐1, NiijukuKatsushika‐kuTokyo125‐8585Japan
- Graduate School of Advanced Science and EngineeringWaseda University3‐4‐1 OkuboShinjuku‐kuTokyo169‐8555Japan
| |
Collapse
|
4
|
Jang K, Kim H, Choi D, Jang S, Chung DK. Staphylococcus aureus utilizes vimentin to internalize human keratinocytes. Front Cell Infect Microbiol 2025; 15:1543186. [PMID: 40061451 PMCID: PMC11885264 DOI: 10.3389/fcimb.2025.1543186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/03/2025] [Indexed: 05/13/2025] Open
Abstract
Introduction Vimentin is an intermediate filamentous cytoskeletal protein involved in cell migration, adhesion, and division. Recent studies have demonstrated that several bacteria and viruses interact with vimentin to facilitate entry and trafficking within eukaryotic cells. However, the relationship between Staphylococcus aureus and vimentin remains unclear. Methods In the current study, we elucidated vimentin expression mechanism in human keratinocytes infected with S. aureus using Western blot (WB), Flow cytometry, Immunofluorescence (IF) staining, utilizing neutralizing antibodies, and small interference (si) RNA, and a vimentin overexpression vector. The physical interaction between vimentin and S. aureus was shown by IF on cell surface, intra- and intercellular space. Results HaCaT cells increased vimentin expression through physical interaction with live S. aureus, and not by heat-killed bacteria or bacterial culture supernatants. The Toll-like receptor (TLR) 2 signaling pathway, which includes interleukin 1 receptor-associated kinase (IRAK) and nuclear factor kappa B (NF-κB)/c-Jun N-terminal kinase (JNK) signaling activation, was involved in S. aureus-mediated vimentin expression. The vimentin protein induced by S. aureus was secreted extracellularly and bound to S. aureus in the culture media. The binding of vimentin to S. aureus accelerated the intracellular infection of HaCaT cells. Discussion Thus, these experiments elucidated the mechanism of vimentin protein expression during S. aureus infection in human skin keratinocytes and revealed the role of vimentin in this process. These findings suggest that vimentin could serve as a potential target for the prevention or treatment of S. aureus infections.
Collapse
Affiliation(s)
- Kyoungok Jang
- Therapeutic Research Group, Antibacterial Resistance Laboratory, Institute Pasteur Korea, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hangeun Kim
- Research and Development Center, Skin Biotechnology Center Co. Ltd., Yongin, Republic of Korea
| | - Dobin Choi
- Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
| | - Soojin Jang
- Therapeutic Research Group, Antibacterial Resistance Laboratory, Institute Pasteur Korea, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Dae-Kyun Chung
- Research and Development Center, Skin Biotechnology Center Co. Ltd., Yongin, Republic of Korea
- Graduate School of Biotechnology, Kyung Hee University, Yongin, Republic of Korea
| |
Collapse
|
5
|
Smoler M, Pennacchietti F, De Rossi MC, Bruno L, Testa I, Levi V. Dynamical organization of vimentin intermediate filaments in living cells revealed by MoNaLISA nanoscopy. Biosci Rep 2025; 45:BSR20241133. [PMID: 39936518 DOI: 10.1042/bsr20241133] [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: 12/23/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 02/13/2025] Open
Abstract
Intermediate filaments are intimately involved in the mechanical behavior of cells. Unfortunately, the resolution of optical microscopy limits our understanding of their organization. Here, we combined nanoscopy, single-filament tracking, and numerical simulations to inspect the dynamical organization of vimentin intermediate filaments in live cells. We show that a higher proportion of peripheral versus perinuclear vimentin pools are constrained in their lateral motion in the seconds time window, probably due to their cross-linking to other cytoskeletal networks. In a longer time scale, active forces become evident and affect similarly both pools of filaments. Our results provide a detailed description of the dynamical organization of the vimentin network in live cells and give some cues on its response to mechanical stimuli.
Collapse
Affiliation(s)
- Mariano Smoler
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, CONICET - Universidad de Buenos Aires, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Francesca Pennacchietti
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - María Cecilia De Rossi
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, CONICET - Universidad de Buenos Aires, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| | - Luciana Bruno
- Facultad de Ciencias Exactas y Naturales, CONICET - Universidad de Buenos Aires, Instituto de Cálculo (IC), Buenos Aires, Argentina
| | - Ilaria Testa
- Department of Applied Physics and Science for Life Laboratory, KTH Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Valeria Levi
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, CONICET - Universidad de Buenos Aires, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, Argentina
| |
Collapse
|
6
|
Huang X, Li Z, Huang Y, Zhang Q, Cui Y, Shi X, Jiu Y. Vimentin intermediate filaments coordinate actin stress fibers and podosomes to determine the extracellular matrix degradation by macrophages. Dev Cell 2025:S1534-5807(25)00036-X. [PMID: 39952241 DOI: 10.1016/j.devcel.2025.01.016] [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: 04/01/2024] [Revised: 09/25/2024] [Accepted: 01/24/2025] [Indexed: 02/17/2025]
Abstract
Macrophages possess the capacity to degrade extracellular matrix (ECM), but the specific roles of different cytoskeletal structures in controlling this process are incompletely understood. Here, we report that the inward flow of actin stress fibers delivers endocytosed ECM for lysosomal elimination, replenishing the pool of enzymes for extracellular ECM hydrolysis in actin-rich podosomes. Vimentin deficiency disrupted the balance between stress fibers and podosomes, impairing ECM degradation through integrin CD11b in THP-1 macrophages. In lung adenocarcinoma patient samples, M2-type macrophages exhibit a tighter podosome organization, surrounded by compact vimentin filaments, than M1-type. In vitro experiments verified that the invasion ability of A549 lung carcinoma cells was enhanced when accompanied by wild type, but not vimentin knockout M2-type THP-1, macrophages. Subcutaneous injections of macrophages and tumor cells in nude mice showed that vimentin in macrophages can reduce tumor collagen fibers. Together, our findings provide insights into the cytoskeletal dynamics governing macrophage ECM degradation.
Collapse
Affiliation(s)
- Xinyi Huang
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhifang Li
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuhan Huang
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Yuquan Road No. 19(A), Shijingshan District, Beijing 100049, China
| | - Qian Zhang
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanqin Cui
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xuemeng Shi
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yaming Jiu
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Yuquan Road No. 19(A), Shijingshan District, Beijing 100049, China.
| |
Collapse
|
7
|
Blob A, Ventzke D, Rölleke U, Nies G, Munk A, Schaedel L, Köster S. Global alignment and local curvature of microtubules in mouse fibroblasts are robust against perturbations of vimentin and actin. SOFT MATTER 2025; 21:641-651. [PMID: 39749806 PMCID: PMC11697242 DOI: 10.1039/d4sm01127a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025]
Abstract
The eukaryotic cytoskeleton is an intricate network of three types of mechanically distinct biopolymers - actin filaments, microtubules and intermediate filaments (IFs). These filamentous networks determine essential cellular functions and properties. Among them, microtubules are important for intracellular transport and establishing cell polarity during migration. Despite their intrinsic stiffness, they exhibit characteristic bending and buckling in cells due to nonthermal forces acting on them. Interactions between cytoskeletal filaments have been found but are complex and diverse with respect to their effect on the mechanical behavior of the filaments and the architecture of networks. We systematically study how actin and vimentin IFs influence the network structure and local bending of microtubules by analyzing fluorescence microscopy images of mouse fibroblasts on protein micropatterns. Our automated analysis averages over large amounts of data to mitigate the effect of the considerable natural variance in biological cell data. We find that the radial orientation of microtubules in circular cells is robust and is established independently of vimentin and actin networks. Observing the local curvature of microtubules, we find highly similar average bending of microtubules in the entire cell regardless of the cytoskeletal surrounding. Small systematic differences cannot be attributed directly to vimentin and actin densities. Our results suggest that, on average, microtubules in unpolarized mouse fibroblasts are unexpectedly independent of the rest of the cytoskeleton in their global network structure and their local curvature.
Collapse
Affiliation(s)
- Anna Blob
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
| | - David Ventzke
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
| | - Ulrike Rölleke
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Germany
| | - Giacomo Nies
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
| | - Axel Munk
- Institute for Mathematical Stochastics, University of Göttingen, Goldschmidtstraße 7, 37077 Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany
| | - Laura Schaedel
- Department of Physics, Center for Biophysics, Saarland University, Campus A2 4, 66123 Saarbrücken, Germany
| | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Friedrich-Hund-Platz 1, 37077 Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany
| |
Collapse
|
8
|
Shiga A, Fujiwara C, Katoh Y, Ito T, Ohnuma-Koyama A, Takahashi N, Harada T. The relationship between spontaneous cystic degeneration and pseudocapillarization in sinusoids in the liver of aged Sprague-Dawley rats. J Toxicol Pathol 2025; 38:27-36. [PMID: 39839718 PMCID: PMC11745498 DOI: 10.1293/tox.2024-0034] [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: 05/15/2024] [Accepted: 08/02/2024] [Indexed: 01/23/2025] Open
Abstract
Cystic degeneration (CD) in the liver is a cyst-like lesion composed of one or more pseudocysts lacking lining cells, occurring spontaneously in rats older than 12 months, with a male predilection. In this study, 32 CDs were identified in 23 out of 104 non-treated, control male Sprague-Dawley rats from two combined chronic toxicity and carcinogenicity studies with agrochemicals. They were examined histologically, histochemically, and immunohistochemically to assess the pathogenesis and pathological significance of CD, focusing on pseudocapillarization in aged rat liver. Pseudocapillarization refers to age-related capillarization of hepatic sinusoids and is distinct from sinusoidal capillarization observed in hepatic cirrhosis. Both CD and pseudocapillarization, characterized by factor VIII-related antigen expression, were primarily noted in the periportal regions of the rat liver. CD areas exhibited enhanced vimentin expression in a diffuse linear pattern in their septa with occasional focal linear α-smooth muscle actin expression and the fluid containing hyaluronic acid accumulated in their lumen that are thought to be formed by hepatocellular apoptosis. These findings suggest a series of reactive changes associated with hepatocellular apoptosis due to pseudocapillarization in the sinusoids. In conclusion, spontaneous CD in rat liver is not a degenerative lesion or cystic enlargement of stellate cells, but a structural abnormality in pre-existing liver tissue resulting from aging-related changes in sinusoidal endothelial cells and hepatocytes. Pseudocapillarization of sinusoids is considered a precursor lesion of CD in the rat liver.
Collapse
Affiliation(s)
- Atsushi Shiga
- The Institute of Environmental Toxicology, 4321
Uchimoriya-machi, Joso-shi, Ibaraki 303-0043, Japan
| | - Chinatsu Fujiwara
- The Institute of Environmental Toxicology, 4321
Uchimoriya-machi, Joso-shi, Ibaraki 303-0043, Japan
| | - Yoshitaka Katoh
- The Institute of Environmental Toxicology, 4321
Uchimoriya-machi, Joso-shi, Ibaraki 303-0043, Japan
| | - Tsuyoshi Ito
- The Institute of Environmental Toxicology, 4321
Uchimoriya-machi, Joso-shi, Ibaraki 303-0043, Japan
| | - Aya Ohnuma-Koyama
- The Institute of Environmental Toxicology, 4321
Uchimoriya-machi, Joso-shi, Ibaraki 303-0043, Japan
| | - Naofumi Takahashi
- The Institute of Environmental Toxicology, 4321
Uchimoriya-machi, Joso-shi, Ibaraki 303-0043, Japan
| | - Takanori Harada
- The Institute of Environmental Toxicology, 4321
Uchimoriya-machi, Joso-shi, Ibaraki 303-0043, Japan
| |
Collapse
|
9
|
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
|
10
|
Jessop E, Young N, Garcia-Del-Valle B, Crusher JT, Obara B, Karakesisoglou I. SIRT2 Inhibition by AGK2 Promotes Perinuclear Cytoskeletal Organisation and Reduces Invasiveness of MDA-MB-231 Triple-Negative Breast Cancer Cells in Confined In Vitro Models. Cells 2024; 13:2023. [PMID: 39682770 PMCID: PMC11639776 DOI: 10.3390/cells13232023] [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: 11/13/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype characterised by the absence of targetable hormone receptors and increased metastatic rates. As nuclear softening strongly contributes to TNBC's enhanced metastatic capacity, increasing the nuclear stiffness of TNBC cells may present a promising therapeutic avenue. Previous evidence has demonstrated the ability of Sirtuin 2 (SIRT2) inhibition to induce cytoskeletal reorganisation, a key factor in regulating nuclear mechanics. Thus, our study aimed to investigate the effect of SIRT2 inhibition on the nuclear mechanics and migratory behaviour of TNBC cells. To achieve this, SIRT2 was pharmacologically inhibited in MDA-MB-231 cells using AGK2, a SIRT2-specific inhibitor. Although SIRT2 inhibition had no effect on LINC complex composition, the AGK2-treated MDA-MB-231 cells displayed more prominent perinuclear organisations of acetylated α-tubulin, vimentin, and F-actin. Additionally, the nuclei of the AGK2-treated MDA-MB-231 cells exhibited greater resistance to collapse under osmotic shock. Scratch-wound assays also revealed that SIRT2 inhibition led to polarity defects in the MDA-MB-231 cells, while in vitro space-restrictive invasion assays highlighted their reduced migratory capacity upon AGK2 treatment. Taken together, our findings suggest that SIRT2 inhibition promotes a perinuclear cytoskeletal organisation in MDA-MB-231 cells, which enhances their nuclear rigidity and impedes their invasion through confined spaces in vitro.
Collapse
Affiliation(s)
- Emily Jessop
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| | - Natalie Young
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| | - Beatriz Garcia-Del-Valle
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| | - Jack T. Crusher
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| | - Boguslaw Obara
- School of Computing, Newcastle University, Newcastle upon Tyne NE4 5TG, UK;
| | - Iakowos Karakesisoglou
- Department of Biosciences, Durham University, Durham DH1 3LE, UK; (E.J.); (N.Y.); (B.G.-D.-V.); (J.T.C.)
| |
Collapse
|
11
|
Srivastava LK, Ehrlicher AJ. Sensing the squeeze: nuclear mechanotransduction in health and disease. Nucleus 2024; 15:2374854. [PMID: 38951951 PMCID: PMC11221475 DOI: 10.1080/19491034.2024.2374854] [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: 02/23/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
The nucleus not only is a repository for DNA but also a center of cellular and nuclear mechanotransduction. From nuclear deformation to the interplay between mechanosensing components and genetic control, the nucleus is poised at the nexus of mechanical forces and cellular function. Understanding the stresses acting on the nucleus, its mechanical properties, and their effects on gene expression is therefore crucial to appreciate its mechanosensitive function. In this review, we examine many elements of nuclear mechanotransduction, and discuss the repercussions on the health of cells and states of illness. By describing the processes that underlie nuclear mechanosensation and analyzing its effects on gene regulation, the review endeavors to open new avenues for studying nuclear mechanics in physiology and diseases.
Collapse
Affiliation(s)
| | - Allen J. Ehrlicher
- Department of Bioengineering, McGill University, Montreal, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada
- Centre for Structural Biology, McGill University, Montreal, Canada
- Department of Mechanical Engineering, McGill University, Montreal, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Canada
| |
Collapse
|
12
|
Zhang Y, Zhu L, Guo Y, Cook C, Ma W, Ran Y, Hu X, Xia Y, Geng S, Liu Y. Clinical and histopathological characteristics of atrophic pigmented dermatofibrosarcoma protuberans: A retrospective study of 14 cases. Heliyon 2024; 10:e39271. [PMID: 39634408 PMCID: PMC11616497 DOI: 10.1016/j.heliyon.2024.e39271] [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: 07/09/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 12/07/2024] Open
Abstract
Background Dermatofibrosarcoma protuberans (DFSP) invades the dermis and subcutaneous tissue. DFSP with both atrophic and pigmentary (AP-DFSP) features is extremely rare and the clinical characteristics remain unknown. Here we aim to characterize the clinical, histopathologic and prognostic features of AP-DFSP. Methods Fourteen cases of patients with AP-DFSP were collected from our institution and published online, including four unreported cases and ten published cases. The clinical appearance, immunohistochemical markers, treatment, and prognosis were analyzed to obtain the clinical and histological features. Results There were six males and eight females with a mean age of 25 years old. The vast majority of lesions appeared in the trunk (10/14, 71.4 %) and limbs (3/14, 21.4 %), whereas a minority involved the infraorbital area (1/14, 7.2 %). The most typical manifestation was a depressed plaque-like lesion with fuchsia and bluish color. Histologically, AP-DFSP harbored both atrophic and pigmented features, presenting with a thinner dermis and intradermal melanin granules. Immunohistochemically, CD34 and vimentin were positive while S100 was negative in tumor tissues. The Ki67 index was less than 10 %. Thirteen of fourteen patients had complete excision surgery and follow-ups showed no local recurrence or distant metastasis. Conclusion Compared to DFSP, AP-DFSP shows more benign clinical and histological features with a good prognosis. Surgical intervention leads to a significant reduction in tumor burden and dramatically increases the likelihood of complete remission.
Collapse
Affiliation(s)
- Yufei Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Longfei Zhu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Ying Guo
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Christopher Cook
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Wenqi Ma
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Yutong Ran
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Xiaoqian Hu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Songmei Geng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| | - Yale Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Shaanxi, China
| |
Collapse
|
13
|
Swoger M, Thanh MTH, Byfield FJ, Dam V, Williamson J, Frank B, Hehnly H, Conway D, Patteson AE. Vimentin molecular linkages with nesprin-3 enhance nuclear deformations by cell geometric constraints. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.621001. [PMID: 39554181 PMCID: PMC11565891 DOI: 10.1101/2024.10.29.621001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The nucleus is the organelle of the cell responsible for controlling protein expression, which has a direct effect on cellular biological functions. Here we show that the cytoskeletal protein vimentin plays an important role in increasing cell-generated forces transmitted to the cell nucleus, resulting in increased nuclear deformations in strongly polarized cells. Using micropatterned substrates to geometrically control cell shape in wild-type and vimentin-null cells, we show vimentin increases polarization and deformation of the cell nucleus. Loss of nesprin-3, which physically couples vimentin to the nuclear envelope, phenotypically copies the loss of vimentin, suggesting vimentin transmits forces to the cell nucleus through direct molecular linkages. Use of a fluorescence resonance energy transfer (FRET) sensor that binds to the nuclear envelope through lamin-A/C suggests vimentin increases the tension on the nuclear envelope. Our results indicate that nuclear shape and deformation can be modified by the vimentin cytoskeleton and its specific crosslinks to the cell nucleus.
Collapse
|
14
|
Chen X, Xu S, Chu B, Guo J, Zhang H, Sun S, Song L, Feng XQ. Applying Spatiotemporal Modeling of Cell Dynamics to Accelerate Drug Development. ACS NANO 2024; 18:29311-29336. [PMID: 39420743 DOI: 10.1021/acsnano.4c12599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Cells act as physical computational programs that utilize input signals to orchestrate molecule-level protein-protein interactions (PPIs), generating and responding to forces, ultimately shaping all of the physiological and pathophysiological behaviors. Genome editing and molecule drugs targeting PPIs hold great promise for the treatments of diseases. Linking genes and molecular drugs with protein-performed cellular behaviors is a key yet challenging issue due to the wide range of spatial and temporal scales involved. Building predictive spatiotemporal modeling systems that can describe the dynamic behaviors of cells intervened by genome editing and molecular drugs at the intersection of biology, chemistry, physics, and computer science will greatly accelerate pharmaceutical advances. Here, we review the mechanical roles of cytoskeletal proteins in orchestrating cellular behaviors alongside significant advancements in biophysical modeling while also addressing the limitations in these models. Then, by integrating generative artificial intelligence (AI) with spatiotemporal multiscale biophysical modeling, we propose a computational pipeline for developing virtual cells, which can simulate and evaluate the therapeutic effects of drugs and genome editing technologies on various cell dynamic behaviors and could have broad biomedical applications. Such virtual cell modeling systems might revolutionize modern biomedical engineering by moving most of the painstaking wet-laboratory effort to computer simulations, substantially saving time and alleviating the financial burden for pharmaceutical industries.
Collapse
Affiliation(s)
- Xindong Chen
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
- BioMap, Beijing 100144, China
| | - Shihao Xu
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Bizhu Chu
- School of Pharmacy, Shenzhen University, Shenzhen 518055, China
- Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jing Guo
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation Research, The First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Huikai Zhang
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Shuyi Sun
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Le Song
- BioMap, Beijing 100144, China
| | - Xi-Qiao Feng
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| |
Collapse
|
15
|
Hjelmeland ME, Lien HE, Berg HF, Woie K, Werner HMJ, Amant F, Haldorsen IS, Trovik J, Krakstad C. Loss of vimentin expression in preoperative biopsies independently predicts poor prognosis, lymph node metastasis and recurrence in endometrial cancer. BJC REPORTS 2024; 2:81. [PMID: 39516342 PMCID: PMC11524127 DOI: 10.1038/s44276-024-00105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Precise preoperative risk classification of endometrial cancer is crucial for treatment decisions. Existing clinical markers often fail to accurately predict lymph node metastasis and recurrence risk. Loss of vimentin expression has emerged as a potential marker for predicting recurrence in low-risk endometrial cancer patients. We assessed whether vimentin expression in preoperative biopsies predicts poor prognosis and lymph node metastasis in a large multicentre cohort. METHODS Vimentin expression was evaluated using immunohistochemistry in 1483 patients diagnosed with endometrial cancer across 14 hospitals in Europe. Expression levels of vimentin were analyzed in conjunction with clinical characteristics for predicting disease-specific survival and lymph node metastases. RESULTS Vimentin loss was significantly associated with aggressive disease and poor survival. Adjusted for clinicopathological variables, vimentin remained independently prognostic with a hazard ratio (HR) of 1.68 (95% CI 1.16-2.42, P = 0.006). Vimentin expression remained independently prognostic in endometrioid endometrial cancer- and FIGO staged 1 patient. Interestingly, vimentin loss independently predicted lymph node metastases, with an HR of 1.83 (95% CI 1.13-2.95, P = 0.014). CONCLUSIONS Loss of vimentin in preoperative biopsies serves as an independent predictor of poor prognosis and lymph node metastases. Incorporating vimentin as a clinical marker can improve risk stratification and treatment decisions.
Collapse
Affiliation(s)
- Marta E Hjelmeland
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Hilde E Lien
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Hege F Berg
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Kathrine Woie
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Henrica M J Werner
- Department of Obstetrics and Gynecology, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
- GROW-Research Institute for Oncology and Reproduction, Maastricht, The Netherlands
| | - Frédéric Amant
- Division Gynecologic Oncology, UZ Leuven, Leuven, Belgium
| | - Ingfrid S Haldorsen
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, Bergen, Norway
| | - Jone Trovik
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Camilla Krakstad
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
16
|
Liu J, Chang X, Manji L, Xu Z, Xiao W. Roles of small peptides encoded by non-coding RNAs in tumor invasion and migration. Front Pharmacol 2024; 15:1442196. [PMID: 39351098 PMCID: PMC11439703 DOI: 10.3389/fphar.2024.1442196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Non-coding RNAs (ncRNAs), which are usually considered not to encode proteins, are widely involved in important activities including signal transduction and cell proliferation. However, recent studies have shown that small peptides encoded by ncRNAs (SPENs) have important roles in the development of malignant tumors. Some SPENs participate in the regulation of skeleton reorganization, intercellular adhesion, signaling and other processes of tumor cells, with effects on the invasive and migratory abilities of the cells. Therefore, SPENs have potential applications as therapeutic targets and biomarkers of malignant tumors. Invasion and migration of malignant tumor cells are the main reasons for poor prognosis of cancer patients and represent the most challenging aspects of treatment of malignant tumors. Currently, the main treatments for tumors include surgery, radiotherapy, targeted drug therapy. Surgery, however, is reserved for early stages of cancer and carries risks and costs. Radiotherapy and targeted therapy have serious side effects. This review describes the mechanisms of SPENs and their roles in tumor invasion and migration, with the aim of providing new targets for tumor diagnosis and treatment.
Collapse
Affiliation(s)
- Jie Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Xiyue Chang
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Laeeqa Manji
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhijie Xu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Wan’an Xiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
17
|
Coelho-Rato LS, Parvanian S, Andrs Salajkova S, Medalia O, Eriksson JE. Intermediate filaments at a glance. J Cell Sci 2024; 137:jcs261386. [PMID: 39206824 DOI: 10.1242/jcs.261386] [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] [Indexed: 09/04/2024] Open
Abstract
Intermediate filaments (IFs) comprise a large family of versatile cytoskeletal proteins, divided into six subtypes with tissue-specific expression patterns. IFs have a wide repertoire of cellular functions, including providing structural support to cells, as well as active roles in mechanical support and signaling pathways. Consequently, defects in IFs are associated with more than 100 diseases. In this Cell Science at a Glance article, we discuss the established classes of IFs and their general features, their functions beyond structural support, and recent advances in the field. We also highlight their involvement in disease and potential use as clinical markers of pathological conditions. Finally, we provide our view on current knowledge gaps and the future directions of the IF field.
Collapse
Affiliation(s)
- Leila S Coelho-Rato
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
| | - Sepideh Parvanian
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Center for Systems Biology, Massachusetts General Hospital Research Institute and Harvard Medical School, Boston, MA 02114, USA
| | - Sarka Andrs Salajkova
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - John E Eriksson
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, 20520 Turku, Finland
- Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, 20520 Turku, Finland
- Euro-Bioimaging ERIC, 20520 Turku, Finland
| |
Collapse
|
18
|
Jawahar A, Vermeil J, Heuvingh J, du Roure O, Piel M. The third dimension of the actin cortex. Curr Opin Cell Biol 2024; 89:102381. [PMID: 38905917 DOI: 10.1016/j.ceb.2024.102381] [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: 02/01/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 06/23/2024]
Abstract
The actin cortex, commonly described as a thin 2-dimensional layer of actin filaments beneath the plasma membrane, is beginning to be recognized as part of a more dynamic and three-dimensional composite material. In this review, we focus on the elements that contribute to the three-dimensional architecture of the actin cortex. We also argue that actin-rich structures such as filopodia and stress fibers can be viewed as specialized integral parts of the 3D actin cortex. This broadens our definition of the cortex, shifting from its simplified characterization as a thin, two-dimensional layer of actin filaments.
Collapse
Affiliation(s)
- Anumita Jawahar
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Université Paris Cité, Sorbonne Université, Paris, France; Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France.
| | - Joseph Vermeil
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Université Paris Cité, Sorbonne Université, Paris, France; Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France
| | - Julien Heuvingh
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Université Paris Cité, Sorbonne Université, Paris, France
| | - Olivia du Roure
- Physique et Mécanique des Milieux Hétérogènes, ESPCI Paris, PSL University, CNRS, Université Paris Cité, Sorbonne Université, Paris, France
| | - Matthieu Piel
- Institut Curie and Institut Pierre Gilles de Gennes, PSL University, CNRS, Paris, France
| |
Collapse
|
19
|
Urbanska M, Guck J. Single-Cell Mechanics: Structural Determinants and Functional Relevance. Annu Rev Biophys 2024; 53:367-395. [PMID: 38382116 DOI: 10.1146/annurev-biophys-030822-030629] [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] [Indexed: 02/23/2024]
Abstract
The mechanical phenotype of a cell determines its ability to deform under force and is therefore relevant to cellular functions that require changes in cell shape, such as migration or circulation through the microvasculature. On the practical level, the mechanical phenotype can be used as a global readout of the cell's functional state, a marker for disease diagnostics, or an input for tissue modeling. We focus our review on the current knowledge of structural components that contribute to the determination of the cellular mechanical properties and highlight the physiological processes in which the mechanical phenotype of the cells is of critical relevance. The ongoing efforts to understand how to efficiently measure and control the mechanical properties of cells will define the progress in the field and drive mechanical phenotyping toward clinical applications.
Collapse
Affiliation(s)
- Marta Urbanska
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jochen Guck
- Max Planck Institute for the Science of Light, Erlangen, Germany; ,
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
20
|
Ho Thanh MT, Poudel A, Ameen S, Carroll B, Wu M, Soman P, Zhang T, Schwarz JM, Patteson AE. Vimentin promotes collective cell migration through collagen networks via increased matrix remodeling and spheroid fluidity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.17.599259. [PMID: 38948855 PMCID: PMC11212918 DOI: 10.1101/2024.06.17.599259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
The intermediate filament (IF) protein vimentin is associated with many diseases with phenotypes of enhanced cellular migration and aggressive invasion through the extracellular matrix (ECM) of tissues, but vimentin's role in in-vivo cell migration is still largely unclear. Vimentin is important for proper cellular adhesion and force generation, which are critical to cell migration; yet the vimentin cytoskeleton also hinders the ability of cells to squeeze through small pores in ECM, resisting migration. To identify the role of vimentin in collective cell migration, we generate spheroids of wide-type and vimentin-null mouse embryonic fibroblasts (mEFs) and embed them in a 3D collagen matrix. We find that loss of vimentin significantly impairs the ability of the spheroid to collectively expand through collagen networks and remodel the collagen network. Traction force analysis reveals that vimentin null spheroids exert less contractile force than their wild-type counterparts. In addition, spheroids made of mEFs with only vimentin unit length filaments (ULFs) exhibit similar behavior as vimentin-null spheroids, suggesting filamentous vimentin is required to promote 3D collective cell migration. We find the vimentin-mediated collective cell expansion is dependent on matrix metalloproteinase (MMP) degradation of the collagen matrix. Further, 3D vertex model simulation of spheroid and embedded ECM indicates that wild-type spheroids behave more fluid-like, enabling more active pulling and reconstructing the surrounding collagen network. Altogether, these results signify that VIF plays a critical role in enhancing migratory persistence in 3D matrix environments through MMP transportation and tissue fluidity.
Collapse
Affiliation(s)
- Minh Tri Ho Thanh
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - Arun Poudel
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Biomedical and Chemical Engineering Department, Syracuse University; Syracuse, New York, USA
| | - Shabeeb Ameen
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - Bobby Carroll
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| | - M Wu
- Department of Biological and Environmental Engineering, Cornell University; Ithaca, New York, USA
| | - Pranav Soman
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Biomedical and Chemical Engineering Department, Syracuse University; Syracuse, New York, USA
| | - Tao Zhang
- Department of Polymer Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - J M Schwarz
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
- Indian Creek Farm, Ithaca, New York, USA
| | - Alison E Patteson
- Physics Department, Syracuse University; Syracuse, New York, USA
- BioInspired Institute, Syracuse University; Syracuse, New York, USA
| |
Collapse
|
21
|
Bae SG, Yin GN, Ock J, Suh JK, Ryu JK, Park J. Single-cell transcriptome analysis of cavernous tissues reveals the key roles of pericytes in diabetic erectile dysfunction. eLife 2024; 12:RP88942. [PMID: 38856719 PMCID: PMC11164535 DOI: 10.7554/elife.88942] [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] [Indexed: 06/11/2024] Open
Abstract
Erectile dysfunction (ED) affects a significant proportion of men aged 40-70 and is caused by cavernous tissue dysfunction. Presently, the most common treatment for ED is phosphodiesterase 5 inhibitors; however, this is less effective in patients with severe vascular disease such as diabetic ED. Therefore, there is a need for development of new treatment, which requires a better understanding of the cavernous microenvironment and cell-cell communications under diabetic condition. Pericytes are vital in penile erection; however, their dysfunction due to diabetes remains unclear. In this study, we performed single-cell RNA sequencing to understand the cellular landscape of cavernous tissues and cell type-specific transcriptional changes in diabetic ED. We found a decreased expression of genes associated with collagen or extracellular matrix organization and angiogenesis in diabetic fibroblasts, chondrocytes, myofibroblasts, valve-related lymphatic endothelial cells, and pericytes. Moreover, the newly identified pericyte-specific marker, Limb Bud-Heart (Lbh), in mouse and human cavernous tissues, clearly distinguishing pericytes from smooth muscle cells. Cell-cell interaction analysis revealed that pericytes are involved in angiogenesis, adhesion, and migration by communicating with other cell types in the corpus cavernosum; however, these interactions were highly reduced under diabetic conditions. Lbh expression is low in diabetic pericytes, and overexpression of LBH prevents erectile function by regulating neurovascular regeneration. Furthermore, the LBH-interacting proteins (Crystallin Alpha B and Vimentin) were identified in mouse cavernous pericytes through LC-MS/MS analysis, indicating that their interactions were critical for maintaining pericyte function. Thus, our study reveals novel targets and insights into the pathogenesis of ED in patients with diabetes.
Collapse
Affiliation(s)
- Seo-Gyeong Bae
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST)GwangjuRepublic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urolog, Inha University School of MedicineIncheonRepublic of Korea
- Program in Biomedical Science & Engineering, Inha UniversityIncheonRepublic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST)GwangjuRepublic of Korea
| |
Collapse
|
22
|
Saldanha R, Ho Thanh MT, Krishnan N, Hehnly H, Patteson A. Vimentin supports cell polarization by enhancing centrosome function and microtubule acetylation. J R Soc Interface 2024; 21:20230641. [PMID: 38835244 DOI: 10.1098/rsif.2023.0641] [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: 11/02/2023] [Accepted: 04/10/2024] [Indexed: 06/06/2024] Open
Abstract
Cell polarity is important for controlling cell shape, motility and cell division processes. Vimentin intermediate filaments are important for cell migration and cell polarization in mesenchymal cells and assembly of vimentin and microtubule networks is dynamically coordinated, but the precise details of how vimentin mediates cell polarity remain unclear. Here, we characterize the effects of vimentin on the structure and function of the centrosome and the stability of microtubule filaments in wild-type and vimentin-null mouse embryonic fibroblasts. We find that vimentin mediates the structure of the pericentriolar material, promotes centrosome-mediated microtubule regrowth and increases the level of stable acetylated microtubules in the cell. Loss of vimentin also impairs centrosome repositioning during cell polarization and migration processes that occur during wound closure. Our results suggest that vimentin modulates centrosome structure and function as well as microtubule network stability, which has important implications for how cells establish proper cell polarization and persistent migration.
Collapse
Affiliation(s)
- Renita Saldanha
- Physics Department, Syracuse University , Syracuse, NY, USA
- BioInspired Institute, Syracuse University , Syracuse, NY, USA
| | - Minh Tri Ho Thanh
- Physics Department, Syracuse University , Syracuse, NY, USA
- BioInspired Institute, Syracuse University , Syracuse, NY, USA
| | - Nikhila Krishnan
- BioInspired Institute, Syracuse University , Syracuse, NY, USA
- Department of Biology, Syracuse University , Syracuse, NY, USA
| | - Heidi Hehnly
- BioInspired Institute, Syracuse University , Syracuse, NY, USA
- Department of Biology, Syracuse University , Syracuse, NY, USA
| | - Alison Patteson
- Physics Department, Syracuse University , Syracuse, NY, USA
- BioInspired Institute, Syracuse University , Syracuse, NY, USA
| |
Collapse
|
23
|
Conboy JP, Istúriz Petitjean I, van der Net A, Koenderink GH. How cytoskeletal crosstalk makes cells move: Bridging cell-free and cell studies. BIOPHYSICS REVIEWS 2024; 5:021307. [PMID: 38840976 PMCID: PMC11151447 DOI: 10.1063/5.0198119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
Cell migration is a fundamental process for life and is highly dependent on the dynamical and mechanical properties of the cytoskeleton. Intensive physical and biochemical crosstalk among actin, microtubules, and intermediate filaments ensures their coordination to facilitate and enable migration. In this review, we discuss the different mechanical aspects that govern cell migration and provide, for each mechanical aspect, a novel perspective by juxtaposing two complementary approaches to the biophysical study of cytoskeletal crosstalk: live-cell studies (often referred to as top-down studies) and cell-free studies (often referred to as bottom-up studies). We summarize the main findings from both experimental approaches, and we provide our perspective on bridging the two perspectives to address the open questions of how cytoskeletal crosstalk governs cell migration and makes cells move.
Collapse
Affiliation(s)
- James P. Conboy
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Irene Istúriz Petitjean
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Anouk van der Net
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Gijsje H. Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| |
Collapse
|
24
|
Alisafaei F, Mandal K, Saldanha R, Swoger M, Yang H, Shi X, Guo M, Hehnly H, Castañeda CA, Janmey PA, Patteson AE, Shenoy VB. Vimentin is a key regulator of cell mechanosensing through opposite actions on actomyosin and microtubule networks. Commun Biol 2024; 7:658. [PMID: 38811770 PMCID: PMC11137025 DOI: 10.1038/s42003-024-06366-4] [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: 11/14/2022] [Accepted: 05/21/2024] [Indexed: 05/31/2024] Open
Abstract
The cytoskeleton is a complex network of interconnected biopolymers consisting of actin filaments, microtubules, and intermediate filaments. These biopolymers work in concert to transmit cell-generated forces to the extracellular matrix required for cell motility, wound healing, and tissue maintenance. While we know cell-generated forces are driven by actomyosin contractility and balanced by microtubule network resistance, the effect of intermediate filaments on cellular forces is unclear. Using a combination of theoretical modeling and experiments, we show that vimentin intermediate filaments tune cell stress by assisting in both actomyosin-based force transmission and reinforcement of microtubule networks under compression. We show that the competition between these two opposing effects of vimentin is regulated by the microenvironment stiffness. These results reconcile seemingly contradictory results in the literature and provide a unified description of vimentin's effects on the transmission of cell contractile forces to the extracellular matrix.
Collapse
Affiliation(s)
- Farid Alisafaei
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Mechanical and Industrial Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Kalpana Mandal
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Medicine and Engineering, University of Pennsylvania, 3340 Smith Walk, Philadelphia, PA, 19104, USA
| | - Renita Saldanha
- Physics Department, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - Maxx Swoger
- Physics Department, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - Haiqian Yang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Xuechen Shi
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Medicine and Engineering, University of Pennsylvania, 3340 Smith Walk, Philadelphia, PA, 19104, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Heidi Hehnly
- Department of Biology, Syracuse University, Syracuse, NY, 13244, USA
| | - Carlos A Castañeda
- Departments of Biology and Chemistry, Syracuse University, Syracuse, NY, 13244, USA
- Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY, 13244, USA
| | - Paul A Janmey
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Medicine and Engineering, University of Pennsylvania, 3340 Smith Walk, Philadelphia, PA, 19104, USA
- Departments of Physiology, and Physics & Astronomy, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alison E Patteson
- Physics Department, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY, 13244, USA
| | - Vivek B Shenoy
- Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
25
|
Moran SJ, Oglietti R, Smith KC, Macosko JC, Holzwarth G, Lyles DS. Mechanisms of active diffusion of vesicular stomatitis virus inclusion bodies and cellular early endosomes in the cytoplasm of mammalian cells. PLoS One 2024; 19:e0290672. [PMID: 38483897 PMCID: PMC10939199 DOI: 10.1371/journal.pone.0290672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
Viral and cellular particles too large to freely diffuse have two different types of mobility in the eukaryotic cell cytoplasm: directed motion mediated by motor proteins moving along cytoskeletal elements with the particle as its load, and motion in random directions mediated by motor proteins interconnecting cytoskeletal elements. The latter motion is referred to as "active diffusion." Mechanisms of directed motion have been extensively studied compared to mechanisms of active diffusion, despite the observation that active diffusion is more common for many viral and cellular particles. Our previous research showed that active diffusion of vesicular stomatitis virus (VSV) ribonucleoproteins (RNPs) in the cytoplasm consists of hopping between traps and that actin filaments and myosin II motors are components of the hop-trap mechanism. This raises the question whether similar mechanisms mediate random motion of larger particles with different physical and biological properties. Live-cell fluorescence imaging and a variational Bayesian analysis used in pattern recognition and machine learning were used to determine the molecular mechanisms of random motion of VSV inclusion bodies and cellular early endosomes. VSV inclusion bodies are membraneless cellular compartments that are the major sites of viral RNA synthesis, and early endosomes are representative of cellular membrane-bound organelles. Like VSV RNPs, inclusion bodies and early endosomes moved from one trapped state to another, but the distance between states was inconsistent with hopping between traps, indicating that the apparent state-to-state movement is mediated by trap movement. Like VSV RNPs, treatment with the actin filament depolymerizing inhibitor latrunculin A increased VSV inclusion body mobility by increasing the size of the traps. In contrast neither treatment with latrunculin A nor depolymerization of microtubules by nocodazole treatment affected the size of traps that confine early endosome mobility, indicating that intermediate filaments are likely major trap components for these cellular organelles.
Collapse
Affiliation(s)
- Steven J. Moran
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Ryan Oglietti
- Department of Biology, Wake Forest University, Winston-Salem, North Carolina, United States of America
| | - Kathleen C. Smith
- Department of Chemistry, Wake Forest University, Winston-Salem, North Carolina, United States of America
| | - Jed C. Macosko
- Department of Physics, Wake Forest University, Winston-Salem, North Carolina, United States of America
| | - George Holzwarth
- Department of Physics, Wake Forest University, Winston-Salem, North Carolina, United States of America
| | - Douglas S. Lyles
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, North Carolina, United States of America
| |
Collapse
|
26
|
Ma T, Liu X, Su H, Shi Q, He Y, Wu F, Gao C, Li K, Liang Z, Zhang D, Zhang X, Hu K, Li S, Wang L, Wang M, Yue S, Hong W, Chen X, Zhang J, Zheng L, Deng X, Wang P, Fan Y. Coupling of Perinuclear Actin Cap and Nuclear Mechanics in Regulating Flow-Induced Yap Spatiotemporal Nucleocytoplasmic Transport. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305867. [PMID: 38161226 PMCID: PMC10953556 DOI: 10.1002/advs.202305867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 11/10/2023] [Indexed: 01/03/2024]
Abstract
Mechanical forces, including flow shear stress, govern fundamental cellular processes by modulating nucleocytoplasmic transport of transcription factors like Yes-associated Protein (YAP). However, the underlying mechanical mechanism remains elusive. In this study, it is reported that unidirectional flow induces biphasic YAP transport with initial nuclear import, followed by nuclear export as actin cap formation and nuclear stiffening. Conversely, pathological oscillatory flow induces slight actin cap formation, nuclear softening, and sustained YAP nuclear localization. To elucidate the disparately YAP spatiotemporal distribution, a 3D mechanochemical model is developed, which integrates flow sensing, cytoskeleton organization, nucleus mechanotransduction, and YAP transport. The results unveiled that despite the significant localized nuclear stress imposed by the actin cap, its inherent stiffness counteracts the dispersed contractile stress exerted by conventional fibers on the nuclear membrane. Moreover, alterations in nuclear stiffness synergistically regulate nuclear deformation, thereby governing YAP transport. Furthermore, by expanding the single-cell model to a collective vertex framework, it is revealed that the irregularities in actin cap formation within individual cells have the potential to induce topological defects and spatially heterogeneous YAP distribution in the cellular monolayer. This work unveils a unified mechanism of flow-induced nucleocytoplasmic transport, providing a linkage between transcription factor localization and mechanical stimulation.
Collapse
Affiliation(s)
- Tianxiang Ma
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Xiao Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Haoran Su
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Qiusheng Shi
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Yuan He
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Fan Wu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Chenxing Gao
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Kexin Li
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Zhuqing Liang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Dongrui Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Xing Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Ke Hu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Shangyu Li
- Biomedical Pioneering Innovation Center (BIOPIC)Peking UniversityBeijing100871China
- Academy for Advanced Interdisciplinary StudiesPeking UniversityBeijing100871China
| | - Li Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Min Wang
- Department of Gynecology and ObstetricsStrategic Support Force Medical CenterBeijing100101China
| | - Shuhua Yue
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Weili Hong
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Xun Chen
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Jing Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Lisha Zheng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Xiaoyan Deng
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Pu Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical EngineeringBeihang UniversityBeijing100083China
- School of Engineering MedicineBeihang UniversityBeijing100083China
| |
Collapse
|
27
|
Chikina AS, Zholudeva AO, Lomakina ME, Kireev II, Dayal AA, Minin AA, Maurin M, Svitkina TM, Alexandrova AY. Plasma Membrane Blebbing Is Controlled by Subcellular Distribution of Vimentin Intermediate Filaments. Cells 2024; 13:105. [PMID: 38201309 PMCID: PMC10778383 DOI: 10.3390/cells13010105] [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: 11/28/2023] [Revised: 12/18/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
The formation of specific cellular protrusions, plasma membrane blebs, underlies the amoeboid mode of cell motility, which is characteristic for free-living amoebae and leukocytes, and can also be adopted by stem and tumor cells to bypass unfavorable migration conditions and thus facilitate their long-distance migration. Not all cells are equally prone to bleb formation. We have previously shown that membrane blebbing can be experimentally induced in a subset of HT1080 fibrosarcoma cells, whereas other cells in the same culture under the same conditions retain non-blebbing mesenchymal morphology. Here we show that this heterogeneity is associated with the distribution of vimentin intermediate filaments (VIFs). Using different approaches to alter the VIF organization, we show that blebbing activity is biased toward cell edges lacking abundant VIFs, whereas the VIF-rich regions of the cell periphery exhibit low blebbing activity. This pattern is observed both in interphase fibroblasts, with and without experimentally induced blebbing, and during mitosis-associated blebbing. Moreover, the downregulation of vimentin expression or displacement of VIFs away from the cell periphery promotes blebbing even in cells resistant to bleb-inducing treatments. Thus, we reveal a new important function of VIFs in cell physiology that involves the regulation of non-apoptotic blebbing essential for amoeboid cell migration and mitosis.
Collapse
Affiliation(s)
- Aleksandra S. Chikina
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow 115478, Russia; (A.S.C.); (A.O.Z.); (M.E.L.)
- Dynamics of Immune Responses Team, INSERM-U1223 Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Anna O. Zholudeva
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow 115478, Russia; (A.S.C.); (A.O.Z.); (M.E.L.)
| | - Maria E. Lomakina
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow 115478, Russia; (A.S.C.); (A.O.Z.); (M.E.L.)
| | - Igor I. Kireev
- Department of Biology and A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, 1 Leninskie Gory, Moscow 119992, Russia;
| | - Alexander A. Dayal
- Institute of Protein Research, Department of Cell Biology, Russian Academy of Sciences, Moscow 119988, Russia; (A.A.D.); (A.A.M.)
| | - Alexander A. Minin
- Institute of Protein Research, Department of Cell Biology, Russian Academy of Sciences, Moscow 119988, Russia; (A.A.D.); (A.A.M.)
| | - Mathieu Maurin
- Institut Curie, PSL Research University, INSERM U932, 26 rue d’Ulm, 75248 Paris, France;
| | - Tatyana M. Svitkina
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Antonina Y. Alexandrova
- N.N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow 115478, Russia; (A.S.C.); (A.O.Z.); (M.E.L.)
| |
Collapse
|
28
|
Schwarz N, Leube RE. Plasticity of cytoplasmic intermediate filament architecture determines cellular functions. Curr Opin Cell Biol 2023; 85:102270. [PMID: 37918274 DOI: 10.1016/j.ceb.2023.102270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/26/2023] [Accepted: 10/02/2023] [Indexed: 11/04/2023]
Abstract
Cytoplasmic intermediate filaments endow cells with mechanical stability. They are subject to changes in morphology and composition if needed. This remodeling encompasses entire cells but can also be restricted to specific intracellular regions. Intermediate filaments thereby support spatially and temporally defined cell type-specific functions. This review focuses on recent advances in our understanding of how intermediate filament dynamics affect the underlying regulatory pathways. We will elaborate on the role of intermediate filaments for the formation and maintenance of surface specializations, cell migration, contractility, organelle positioning, nucleus protection, stress responses and axonal conduction velocity. Together, the selected examples highlight the modulatory role of intermediate filament plasticity for multiple cellular functions.
Collapse
Affiliation(s)
- Nicole Schwarz
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, Wendlingweg 2, 52074 Aachen, Germany.
| |
Collapse
|
29
|
Gu Q, Shanahan L, Hart JW, Belser S, Shofer N, Atatüre M, Knowles HS. Simultaneous Nanorheometry and Nanothermometry Using Intracellular Diamond Quantum Sensors. ACS NANO 2023; 17:20034-20042. [PMID: 37791968 PMCID: PMC10604098 DOI: 10.1021/acsnano.3c05285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 09/15/2023] [Indexed: 10/05/2023]
Abstract
The viscoelasticity of the cytoplasm plays a critical role in cell morphology, cell division, and intracellular transport. Viscoelasticity is also interconnected with other biophysical properties, such as temperature, which is known to influence cellular bioenergetics. Probing the connections between intracellular temperature and cytoplasmic viscoelasticity provides an exciting opportunity for the study of biological phenomena, such as metabolism and disease progression. The small length scales and transient nature of changes in these parameters combined with their complex interdependencies pose a challenge for biosensing tools, which are often limited to a single readout modality. Here, we present a dual-mode quantum sensor capable of performing simultaneous nanoscale thermometry and rheometry in dynamic cellular environments. We use nitrogen-vacancy centers in diamond nanocrystals as biocompatible sensors for in vitro measurements. We combine subdiffraction resolution single-particle tracking in a fluidic environment with optically detected magnetic resonance spectroscopy to perform simultaneous sensing of viscoelasticity and temperature. We use our sensor to demonstrate probing of the temperature-dependent viscoelasticity in complex media at the nanoscale. We then investigate the interplay between intracellular forces and the cytoplasmic rheology in live cells. Finally, we identify different rheological regimes and reveal evidence of active trafficking and details of the nanoscale viscoelasticity of the cytoplasm.
Collapse
Affiliation(s)
| | | | | | - Sophia Belser
- Cavendish Laboratory, University of Cambridge, JJ Thompson Avenue, Cambridge CB3 0HE, United
Kingdom
| | - Noah Shofer
- Cavendish Laboratory, University of Cambridge, JJ Thompson Avenue, Cambridge CB3 0HE, United
Kingdom
| | - Mete Atatüre
- Cavendish Laboratory, University of Cambridge, JJ Thompson Avenue, Cambridge CB3 0HE, United
Kingdom
| | - Helena S. Knowles
- Cavendish Laboratory, University of Cambridge, JJ Thompson Avenue, Cambridge CB3 0HE, United
Kingdom
| |
Collapse
|
30
|
Jebane C, Varlet AA, Karnat M, Hernandez- Cedillo LM, Lecchi A, Bedu F, Desgrouas C, Vigouroux C, Vantyghem MC, Viallat A, Rupprecht JF, Helfer E, Badens C. Enhanced cell viscosity: A new phenotype associated with lamin A/C alterations. iScience 2023; 26:107714. [PMID: 37701573 PMCID: PMC10494210 DOI: 10.1016/j.isci.2023.107714] [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: 05/03/2023] [Revised: 07/13/2023] [Accepted: 08/22/2023] [Indexed: 09/14/2023] Open
Abstract
Lamin A/C is a well-established key contributor to nuclear stiffness and its role in nucleus mechanical properties has been extensively studied. However, its impact on whole-cell mechanics has been poorly addressed, particularly concerning measurable physical parameters. In this study, we combined microfluidic experiments with theoretical analyses to quantitatively estimate the whole-cell mechanical properties. This allowed us to characterize the mechanical changes induced in cells by lamin A/C alterations and prelamin A accumulation resulting from atazanavir treatment or lipodystrophy-associated LMNA R482W pathogenic variant. Our results reveal a distinctive increase in long-time viscosity as a signature of cells affected by lamin A/C alterations. Furthermore, they show that the whole-cell response to mechanical stress is driven not only by the nucleus but also by the nucleo-cytoskeleton links and the microtubule network. The enhanced cell viscosity assessed with our microfluidic assay could serve as a valuable diagnosis marker for lamin-related diseases.
Collapse
Affiliation(s)
- Cécile Jebane
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | | | - Marc Karnat
- Aix Marseille Univ, Université de Toulon, CNRS, CPT, Turing Centre for Living Systems, Marseille, France
| | | | | | | | | | - Corinne Vigouroux
- Assistance Publique–Hôpitaux de Paris (AP-HP), Saint-Antoine Hospital, National Reference Centre for Rares diseases of Insulin-Secretion and Insulin-Sensitivity (PRISIS), Department of Endocrinology, Paris, France
- Sorbonne University, Saint-Antoine Research Centre, Inserm UMR_S938, Institute of Cardiometabolism and Nutrition, Paris, France
| | - Marie-Christine Vantyghem
- Endocrinology, Diabetology and Metabolism Department, Inserm U1190, EGID, Lille University Hospital, Lille, France
| | - Annie Viallat
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | - Jean-François Rupprecht
- Aix Marseille Univ, Université de Toulon, CNRS, CPT, Turing Centre for Living Systems, Marseille, France
| | - Emmanuèle Helfer
- Aix Marseille Univ, CNRS, CINAM, Turing Centre for Living Systems, Marseille, France
| | - Catherine Badens
- Aix Marseille Univ, INSERM, MMG, Marseille, France
- AP-HM, Laboratoire de Biochimie, Marseille, France
| |
Collapse
|
31
|
Di Conza G, Barbaro F, Zini N, Spaletta G, Remaggi G, Elviri L, Mosca S, Caravelli S, Mosca M, Toni R. Woven bone formation and mineralization by rat mesenchymal stromal cells imply increased expression of the intermediate filament desmin. Front Endocrinol (Lausanne) 2023; 14:1234569. [PMID: 37732119 PMCID: PMC10507407 DOI: 10.3389/fendo.2023.1234569] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/07/2023] [Indexed: 09/22/2023] Open
Abstract
Background Disordered and hypomineralized woven bone formation by dysfunctional mesenchymal stromal cells (MSCs) characterize delayed fracture healing and endocrine -metabolic bone disorders like fibrous dysplasia and Paget disease of bone. To shed light on molecular players in osteoblast differentiation, woven bone formation, and mineralization by MSCs we looked at the intermediate filament desmin (DES) during the skeletogenic commitment of rat bone marrow MSCs (rBMSCs), where its bone-related action remains elusive. Results Monolayer cultures of immunophenotypically- and morphologically - characterized, adult male rBMSCs showed co-localization of desmin (DES) with vimentin, F-actin, and runx2 in all cell morphotypes, each contributing to sparse and dense colonies. Proteomic analysis of these cells revealed a topologically-relevant interactome, focused on cytoskeletal and related enzymes//chaperone/signalling molecules linking DES to runx2 and alkaline phosphatase (ALP). Osteogenic differentiation led to mineralized woven bone nodules confined to dense colonies, significantly smaller and more circular with respect to controls. It significantly increased also colony-forming efficiency and the number of DES-immunoreactive dense colonies, and immunostaining of co-localized DES/runx-2 and DES/ALP. These data confirmed pre-osteoblastic and osteoblastic differentiation, woven bone formation, and mineralization, supporting DES as a player in the molecular pathway leading to the osteogenic fate of rBMSCs. Conclusion Immunocytochemical and morphometric studies coupled with proteomic and bioinformatic analysis support the concept that DES may act as an upstream signal for the skeletogenic commitment of rBMSCs. Thus, we suggest that altered metabolism of osteoblasts, woven bone, and mineralization by dysfunctional BMSCs might early be revealed by changes in DES expression//levels. Non-union fractures and endocrine - metabolic bone disorders like fibrous dysplasia and Paget disease of bone might take advantage of this molecular evidence for their early diagnosis and follow-up.
Collapse
Affiliation(s)
- Giusy Di Conza
- Department of Medicine and Surgery - DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine - BIOMED, University of Parma, Parma, Italy
| | - Fulvio Barbaro
- Department of Medicine and Surgery - DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine - BIOMED, University of Parma, Parma, Italy
| | - Nicoletta Zini
- Unit of Bologna, National Research Council of Italy (CNR) Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Giulia Spaletta
- Department of Statistical Sciences, University of Bologna, Bologna, Italy
| | - Giulia Remaggi
- Food and Drug Department, University of Parma, Parma, Italy
| | - Lisa Elviri
- Food and Drug Department, University of Parma, Parma, Italy
| | - Salvatore Mosca
- Course on Disorders of the Locomotor System, Fellow Program in Orthopaedics and Traumatology, University Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Caravelli
- II Clinic of Orthopedic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Massimiliano Mosca
- II Clinic of Orthopedic and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Roberto Toni
- Department of Medicine and Surgery - DIMEC, Unit of Biomedical, Biotechnological and Translational Sciences (S.BI.BI.T.), Laboratory of Regenerative Morphology and Bioartificial Structures (Re.Mo.Bio.S.), and Museum and Historical Library of Biomedicine - BIOMED, University of Parma, Parma, Italy
- Endocrinology, Diabetes, and Nutrition Disorders Outpatient Clinic, Osteoporosis, Nutrition, Endocrinology, and Innovative Therapies (OSTEONET) Unit, Galliera Medical Center (GMC), San Venanzio di Galliera, BO, Italy
- Section IV - Medical Sciences, Academy of Sciences of the Institute of Bologna, Bologna, Italy
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Tufts Medical Center - Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
32
|
Zhu Y, Zhang M, Sun Q, Wang X, Li X, Li Q. Advanced Mechanical Testing Technologies at the Cellular Level: The Mechanisms and Application in Tissue Engineering. Polymers (Basel) 2023; 15:3255. [PMID: 37571149 PMCID: PMC10422338 DOI: 10.3390/polym15153255] [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: 07/11/2023] [Revised: 07/24/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Mechanics, as a key physical factor which affects cell function and tissue regeneration, is attracting the attention of researchers in the fields of biomaterials, biomechanics, and tissue engineering. The macroscopic mechanical properties of tissue engineering scaffolds have been studied and optimized based on different applications. However, the mechanical properties of the overall scaffold materials are not enough to reveal the mechanical mechanism of the cell-matrix interaction. Hence, the mechanical detection of cell mechanics and cellular-scale microenvironments has become crucial for unraveling the mechanisms which underly cell activities and which are affected by physical factors. This review mainly focuses on the advanced technologies and applications of cell-scale mechanical detection. It summarizes the techniques used in micromechanical performance analysis, including atomic force microscope (AFM), optical tweezer (OT), magnetic tweezer (MT), and traction force microscope (TFM), and analyzes their testing mechanisms. In addition, the application of mechanical testing techniques to cell mechanics and tissue engineering scaffolds, such as hydrogels and porous scaffolds, is summarized and discussed. Finally, it highlights the challenges and prospects of this field. This review is believed to provide valuable insights into micromechanics in tissue engineering.
Collapse
Affiliation(s)
- Yingxuan Zhu
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Mengqi Zhang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qingqing Sun
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaofeng Wang
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaomeng Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| | - Qian Li
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou 450001, China
- National Center for International Joint Research of Micro-nano Moulding Technology, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
33
|
Lee S, Jiao M, Zhang Z, Yu Y. Nanoparticles for Interrogation of Cell Signaling. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2023; 16:333-351. [PMID: 37314874 PMCID: PMC10627408 DOI: 10.1146/annurev-anchem-092822-085852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cell functions rely on signal transduction-the cascades of molecular interactions and biochemical reactions that relay extracellular signals to the cell interior. Dissecting principles governing the signal transduction process is critical for the fundamental understanding of cell physiology and the development of biomedical interventions. The complexity of cell signaling is, however, beyond what is accessible by conventional biochemistry assays. Thanks to their unique physical and chemical properties, nanoparticles (NPs) have been increasingly used for the quantitative measurement and manipulation of cell signaling. Even though research in this area is still in its infancy, it has the potential to yield new, paradigm-shifting knowledge of cell biology and lead to biomedical innovations. To highlight this importance, we summarize in this review studies that pioneered the development and application of NPs for cell signaling, from quantitative measurements of signaling molecules to spatiotemporal manipulation of cell signal transduction.
Collapse
Affiliation(s)
- Seonik Lee
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA;
| | - Mengchi Jiao
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA;
| | - Zihan Zhang
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA;
| | - Yan Yu
- Department of Chemistry, Indiana University, Bloomington, Indiana, USA;
| |
Collapse
|
34
|
Higgins G, Higgins F, Peres J, Lang DM, Abdalrahman T, Zaman MH, Prince S, Franz T. Intracellular mechanics and TBX3 expression jointly dictate the spreading mode of melanoma cells in 3D environments. Exp Cell Res 2023; 428:113633. [PMID: 37172754 DOI: 10.1016/j.yexcr.2023.113633] [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: 03/21/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Cell stiffness and T-box transcription factor 3 (TBX3) expression have been identified as biomarkers of melanoma metastasis in 2D environments. This study aimed to determine how mechanical and biochemical properties of melanoma cells change during cluster formation in 3D environments. Vertical growth phase (VGP) and metastatic (MET) melanoma cells were embedded in 3D collagen matrices of 2 and 4 mg/ml collagen concentrations, representing low and high matrix stiffness. Mitochondrial fluctuation, intracellular stiffness, and TBX3 expression were quantified before and during cluster formation. In isolated cells, mitochondrial fluctuation decreased and intracellular stiffness increased with increase in disease stage from VGP to MET and increased matrix stiffness. TBX3 was highly expressed in soft matrices but diminished in stiff matrices for VGP and MET cells. Cluster formation of VGP cells was excessive in soft matrices but limited in stiff matrices, whereas for MET cells it was limited in soft and stiff matrices. In soft matrices, VGP cells did not change the intracellular properties, whereas MET cells exhibited increased mitochondrial fluctuation and decreased TBX3 expression. In stiff matrices, mitochondrial fluctuation and TBX3 expression increased in VGP and MET, and intracellular stiffness increased in VGP but decreased in MET cells. The findings suggest that soft extracellular environments are more favourable for tumour growth, and high TBX3 levels mediate collective cell migration and tumour growth in the earlier VGP disease stage but play a lesser role in the later metastatic stage of melanoma.
Collapse
Affiliation(s)
- Ghodeejah Higgins
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Faatiemah Higgins
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Jade Peres
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Dirk M Lang
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Tamer Abdalrahman
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Muhammad H Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Sharon Prince
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Observatory, South Africa
| | - Thomas Franz
- Biomedical Engineering Research Centre, Division of Biomedical Engineering, Department of Human Biology, University of Cape Town, Observatory, South Africa; Bioengineering Science Research Group, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
35
|
Mahmoudian RA, Akhlaghipour I, Lotfi M, Shahidsales S, Moghbeli M. Circular RNAs as the pivotal regulators of epithelial-mesenchymal transition in gastrointestinal tumor cells. Pathol Res Pract 2023; 245:154472. [PMID: 37087995 DOI: 10.1016/j.prp.2023.154472] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 04/25/2023]
Abstract
Gastrointestinal (GI) cancers, as the most common human malignancies are always considered one of the most important health challenges in the world. Late diagnosis in advanced tumor stages is one of the main reasons for the high mortality rate and treatment failure in these patients. Therefore, investigating the molecular pathways involved in GI tumor progression is required to introduce the efficient markers for the early tumor diagnosis. Epithelial-mesenchymal transition (EMT) is one of the main cellular mechanisms involved in the GI tumor metastasis. Non-coding RNAs (ncRNAs) are one of the main regulatory factors in EMT process. Circular RNAs (circRNAs) are a group of covalently closed loop ncRNAs that have higher stability in body fluids compared with other ncRNAs. Considering the importance of circRNAs in regulation of EMT process, in the present review we discussed the role of circRNAs in EMT process during GI tumor invasion. It has been reported that circRNAs mainly affect the EMT process through the regulation of EMT-specific transcription factors and signaling pathways such as WNT, PI3K/AKT, TGF-β, and MAPK. This review can be an effective step in introducing a circRNA/EMT based diagnostic panel marker for the early tumor detection among GI cancer patients.
Collapse
Affiliation(s)
- Reihaneh Alsadat Mahmoudian
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Lotfi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
36
|
Fernández Casafuz AB, De Rossi MC, Bruno L. Mitochondrial cellular organization and shape fluctuations are differentially modulated by cytoskeletal networks. Sci Rep 2023; 13:4065. [PMID: 36906690 PMCID: PMC10008531 DOI: 10.1038/s41598-023-31121-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/07/2023] [Indexed: 03/13/2023] Open
Abstract
The interactions between mitochondria and the cytoskeleton have been found to alter mitochondrial function; however, the mechanisms underlying this phenomenon are largely unknown. Here, we explored how the integrity of the cytoskeleton affects the cellular organization, morphology and mobility of mitochondria in Xenopus laevis melanocytes. Cells were imaged in control condition and after different treatments that selectively affect specific cytoskeletal networks (microtubules, F-actin and vimentin filaments). We observed that mitochondria cellular distribution and local orientation rely mostly on microtubules, positioning these filaments as the main scaffolding of mitochondrial organization. We also found that cytoskeletal networks mold mitochondria shapes in distinct ways: while microtubules favor more elongated organelles, vimentin and actin filaments increase mitochondrial bending, suggesting the presence of mechanical interactions between these filaments and mitochondria. Finally, we identified that microtubule and F-actin networks play opposite roles in mitochondria shape fluctuations and mobility, with microtubules transmitting their jittering to the organelles and F-actin restricting the organelles motion. All our results support that cytoskeleton filaments interact mechanically with mitochondria and transmit forces to these organelles molding their movements and shapes.
Collapse
Affiliation(s)
- Agustina Belén Fernández Casafuz
- CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Cálculo (IC), Buenos Aires, 1428, Argentina
| | - María Cecilia De Rossi
- CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Instituto de Química Biológica (IQUIBICEN), Buenos Aires, 1428, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| | - Luciana Bruno
- CONICET-Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Instituto de Cálculo (IC), Buenos Aires, 1428, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.
| |
Collapse
|
37
|
Shintani T, Suzuki R, Takeuchi Y, Shirasawa T, Noda M. Deletion or inhibition of PTPRO prevents ectopic fat accumulation and induces healthy obesity with markedly reduced systemic inflammation. Life Sci 2023; 313:121292. [PMID: 36535401 DOI: 10.1016/j.lfs.2022.121292] [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: 11/02/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
AIMS Chronic inflammation plays crucial roles in obesity-induced metabolic diseases. Protein tyrosine phosphatase receptor type O (PTPRO) is a member of the R3 subfamily of receptor-like protein tyrosine phosphatases. We previously suggested a role for PTPRO in the inactivation of the insulin receptor. The present study aimed to elucidate the involvement of PTPRO in the control of glucose and lipid metabolism as well as in obesity-induced systemic inflammation. MATERIALS AND METHODS Lipid accumulation in adipose tissue and the liver, the expression of inflammatory cytokines, and insulin resistance associated with systemic inflammation were investigated in hyper-obese Ptpro-KO mice by feeding a high-fat/high-sucrose diet (HFHSD). The effects of the administration of AKB9778, a specific inhibitor of PTPRO, to ob/ob mice and cultured 3T3-L1 preadipocyte cells were also examined. KEY FINDINGS Ptpro was highly expressed in visceral white adipose tissue and macrophages. Ptpro-KO mice fed HFHSD were hyper-obese, but did not have ectopic fat accumulation in the liver, dysfunctional lipid and glucose homeostasis, systemic inflammation, or insulin resistance. The administration of AKB9778 reproduced "the healthy obese phenotypes" of Ptpro-KO mice in highly obese ob/ob mice. Furthermore, the inhibition of PTPRO promoted the growth of lipid droplets in adipocytes through an increase in the phosphorylation of Tyr(117) in vimentin. SIGNIFICANCE Healthy systemic conditions with the attenuation of inflammation in hyper-obese Ptpro-KO mice were associated with the expansion of adipose tissue and low activation of NF-κb. Therefore, PTPRO may be a promising target to ameliorate hepatic steatosis and metabolic dysfunction.
Collapse
Affiliation(s)
- Takafumi Shintani
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8503, Japan; Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan
| | - Ryoko Suzuki
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan
| | - Yasushi Takeuchi
- Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan
| | | | - Masaharu Noda
- Homeostatic Mechanism Research Unit, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa 226-8503, Japan; Division of Molecular Neurobiology, National Institute for Basic Biology, Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
38
|
RANDHAWA AAYUSHI, DEB DUTTA SAYAN, GANGULY KEYA, V. PATIL TEJAL, LUTHFIKASARI RACHMI, LIM KITAEK. Understanding cell-extracellular matrix interactions for topology-guided tissue regeneration. BIOCELL 2023. [DOI: 10.32604/biocell.2023.026217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
39
|
The Forces behind Directed Cell Migration. BIOPHYSICA 2022. [DOI: 10.3390/biophysica2040046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Directed cell migration is an essential building block of life, present when an embryo develops, a dendritic cell migrates toward a lymphatic vessel, or a fibrotic organ fails to restore its normal parenchyma. Directed cell migration is often guided by spatial gradients in a physicochemical property of the cell microenvironment, such as a gradient in chemical factors dissolved in the medium or a gradient in the mechanical properties of the substrate. Single cells and tissues sense these gradients, establish a back-to-front polarity, and coordinate the migration machinery accordingly. Central to these steps we find physical forces. In some cases, these forces are integrated into the gradient sensing mechanism. Other times, they transmit information through cells and tissues to coordinate a collective response. At any time, they participate in the cellular migratory system. In this review, we explore the role of physical forces in gradient sensing, polarization, and coordinating movement from single cells to multicellular collectives. We use the framework proposed by the molecular clutch model and explore to what extent asymmetries in the different elements of the clutch can lead to directional migration.
Collapse
|
40
|
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
|
41
|
Kuburich NA, den Hollander P, Pietz JT, Mani SA. Vimentin and cytokeratin: Good alone, bad together. Semin Cancer Biol 2022; 86:816-826. [PMID: 34953942 PMCID: PMC9213573 DOI: 10.1016/j.semcancer.2021.12.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/26/2021] [Accepted: 12/14/2021] [Indexed: 01/27/2023]
Abstract
The cytoskeleton plays an integral role in maintaining the integrity of epithelial cells. Epithelial cells primarily employ cytokeratin in their cytoskeleton, whereas mesenchymal cells use vimentin. During the epithelial-mesenchymal transition (EMT), cytokeratin-positive epithelial cells begin to express vimentin. EMT induces stem cell properties and drives metastasis, chemoresistance, and tumor relapse. Most studies of the functions of cytokeratin and vimentin have relied on the use of either epithelial or mesenchymal cell types. However, it is important to understand how these two cytoskeleton intermediate filaments function when co-expressed in cells undergoing EMT. Here, we discuss the individual and shared functions of cytokeratin and vimentin that coalesce during EMT and how alterations in intermediate filament expression influence carcinoma progression.
Collapse
Affiliation(s)
- Nick A Kuburich
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Petra den Hollander
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Jordan T Pietz
- Department of Creative Services, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States.
| |
Collapse
|
42
|
Shaebani MR, Stankevicins L, Vesperini D, Urbanska M, Flormann DAD, Terriac E, Gad AKB, Cheng F, Eriksson JE, Lautenschläger F. Effects of vimentin on the migration, search efficiency, and mechanical resilience of dendritic cells. Biophys J 2022; 121:3950-3961. [PMID: 36056556 PMCID: PMC9675030 DOI: 10.1016/j.bpj.2022.08.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/20/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Dendritic cells use amoeboid migration to pass through narrow passages in the extracellular matrix and confined tissue in search for pathogens and to reach the lymph nodes and alert the immune system. Amoeboid migration is a migration mode that, instead of relying on cell adhesion, is based on mechanical resilience and friction. To better understand the role of intermediate filaments in ameboid migration, we studied the effects of vimentin on the migration of dendritic cells. We show that the lymph node homing of vimentin-deficient cells is reduced in our in vivo experiments in mice. Lack of vimentin also reduces the cell stiffness, the number of migrating cells, and the migration speed in vitro in both 1D and 2D confined environments. Moreover, we find that lack of vimentin weakens the correlation between directional persistence and migration speed. Thus, vimentin-expressing dendritic cells move faster in straighter lines. Our numerical simulations of persistent random search in confined geometries verify that the reduced migration speed and the weaker correlation between the speed and direction of motion result in longer search times to find regularly located targets. Together, these observations show that vimentin enhances the ameboid migration of dendritic cells, which is relevant for the efficiency of their random search for pathogens.
Collapse
Affiliation(s)
- M Reza Shaebani
- Department of Theoretical Physics, Saarland University, Saarbrücken, Germany; Centre for Biophysics, Saarland University, Saarbrücken, Germany
| | - Luiza Stankevicins
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Doriane Vesperini
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Marta Urbanska
- Biotechnology Centre, Centre for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Daniel A D Flormann
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Emmanuel Terriac
- Department of Experimental Physics, Saarland University, Saarbrücken, Germany
| | - Annica K B Gad
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom; Centro de Química da Madeira, Universidade da Madeira, Funchal, Portugal
| | - Fang Cheng
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, China
| | - John E Eriksson
- Cell Biology, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland; Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Franziska Lautenschläger
- Centre for Biophysics, Saarland University, Saarbrücken, Germany; Department of Experimental Physics, Saarland University, Saarbrücken, Germany.
| |
Collapse
|
43
|
Sivagurunathan S, Vahabikashi A, Yang H, Zhang J, Vazquez K, Rajasundaram D, Politanska Y, Abdala-Valencia H, Notbohm J, Guo M, Adam SA, Goldman RD. Expression of vimentin alters cell mechanics, cell-cell adhesion, and gene expression profiles suggesting the induction of a hybrid EMT in human mammary epithelial cells. Front Cell Dev Biol 2022; 10:929495. [PMID: 36200046 PMCID: PMC9527304 DOI: 10.3389/fcell.2022.929495] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Vimentin is a Type III intermediate filament (VIF) cytoskeletal protein that regulates the mechanical and migratory behavior of cells. Its expression is considered to be a marker for the epithelial to mesenchymal transition (EMT) that takes place in tumor metastasis. However, the molecular mechanisms regulated by the expression of vimentin in the EMT remain largely unexplored. We created MCF7 epithelial cell lines expressing vimentin from a cumate-inducible promoter to address this question. When vimentin expression was induced in these cells, extensive cytoplasmic VIF networks were assembled accompanied by changes in the organization of the endogenous keratin intermediate filament networks and disruption of desmosomes. Significant reductions in intercellular forces by the cells expressing VIFs were measured by quantitative monolayer traction force and stress microscopy. In contrast, laser trapping micro-rheology revealed that the cytoplasm of MCF7 cells expressing VIFs was stiffer than the uninduced cells. Vimentin expression activated transcription of genes involved in pathways responsible for cell migration and locomotion. Importantly, the EMT related transcription factor TWIST1 was upregulated only in wild type vimentin expressing cells and not in cells expressing a mutant non-polymerized form of vimentin, which only formed unit length filaments (ULF). Taken together, our results suggest that vimentin expression induces a hybrid EMT correlated with the upregulation of genes involved in cell migration.
Collapse
Affiliation(s)
- Suganya Sivagurunathan
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Amir Vahabikashi
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Haiqian Yang
- Department of Mechanical Engineering , Massachusetts Institute of Technology , Cambridge , MA, United States
| | - Jun Zhang
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
| | - Kelly Vazquez
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yuliya Politanska
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Hiam Abdala-Valencia
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jacob Notbohm
- Biophysics Program, University of Wisconsin-Madison, Madison, WI, United States
- Department of Engineering Physics, University of Wisconsin-Madison, Madison, WI, United States
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Ming Guo
- Department of Mechanical Engineering , Massachusetts Institute of Technology , Cambridge , MA, United States
| | - Stephen A Adam
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Robert D Goldman
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
44
|
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
|
45
|
Li Y, Wong IY, Guo M. Reciprocity of Cell Mechanics with Extracellular Stimuli: Emerging Opportunities for Translational Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107305. [PMID: 35319155 PMCID: PMC9463119 DOI: 10.1002/smll.202107305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/20/2022] [Indexed: 06/14/2023]
Abstract
Human cells encounter dynamic mechanical cues in healthy and diseased tissues, which regulate their molecular and biophysical phenotype, including intracellular mechanics as well as force generation. Recent developments in bio/nanomaterials and microfluidics permit exquisitely sensitive measurements of cell mechanics, as well as spatiotemporal control over external mechanical stimuli to regulate cell behavior. In this review, the mechanobiology of cells interacting bidirectionally with their surrounding microenvironment, and the potential relevance for translational medicine are considered. Key fundamental concepts underlying the mechanics of living cells as well as the extracelluar matrix are first introduced. Then the authors consider case studies based on 1) microfluidic measurements of nonadherent cell deformability, 2) cell migration on micro/nano-topographies, 3) traction measurements of cells in three-dimensional (3D) matrix, 4) mechanical programming of organoid morphogenesis, as well as 5) active mechanical stimuli for potential therapeutics. These examples highlight the promise of disease diagnosis using mechanical measurements, a systems-level understanding linking molecular with biophysical phenotype, as well as therapies based on mechanical perturbations. This review concludes with a critical discussion of these emerging technologies and future directions at the interface of engineering, biology, and medicine.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, Hubei, 430074, China
| | - Ian Y Wong
- School of Engineering, Center for Biomedical Engineering, Joint Program in Cancer Biology, Brown University, 184 Hope St Box D, Providence, RI, 02912, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
46
|
Català-Castro F, Schäffer E, Krieg M. Exploring cell and tissue mechanics with optical tweezers. J Cell Sci 2022; 135:jcs259355. [PMID: 35942913 DOI: 10.1242/jcs.259355] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cellular and tissue biosystems emerge from the assembly of their constituent molecules and obtain a set of specific material properties. To measure these properties and understand how they influence cellular function is a central goal of mechanobiology. From a bottoms-up, physics or engineering point-of-view, such systems are a composition of basic mechanical elements. However, the sheer number and dynamic complexity of them, including active molecular machines and their emergent properties, makes it currently intractable to calculate how biosystems respond to forces. Because many diseases result from an aberrant mechanotransduction, it is thus essential to measure this response. Recent advances in the technology of optical tweezers have broadened their scope from single-molecule applications to measurements inside complex cellular environments, even within tissues and animals. Here, we summarize the basic optical trapping principles, implementations and calibration procedures that enable force measurements using optical tweezers directly inside cells of living animals, in combination with complementary techniques. We review their versatility to manipulate subcellular organelles and measure cellular frequency-dependent mechanics in the piconewton force range from microseconds to hours. As an outlook, we address future challenges to fully unlock the potential of optical tweezers for mechanobiology.
Collapse
Affiliation(s)
- Frederic Català-Castro
- Neurophotonics and Mechanical Systems Biology, ICFO, Institut de Ciències Fotòniques, 08860 Castelldefels, Spain
| | - Erik Schäffer
- Cellular Nanoscience, ZMBP, University of Tübingen, 72076 Tübingen, Germany
| | - Michael Krieg
- Neurophotonics and Mechanical Systems Biology, ICFO, Institut de Ciències Fotòniques, 08860 Castelldefels, Spain
| |
Collapse
|
47
|
Verwei HN, Lee G, Leech G, Petitjean II, Koenderink GH, Robertson-Anderson RM, McGorty RJ. Quantifying Cytoskeleton Dynamics Using Differential Dynamic Microscopy. J Vis Exp 2022:10.3791/63931. [PMID: 35781524 PMCID: PMC10398790 DOI: 10.3791/63931] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023] Open
Abstract
Cells can crawl, self-heal, and tune their stiffness due to their remarkably dynamic cytoskeleton. As such, reconstituting networks of cytoskeletal biopolymers may lead to a host of active and adaptable materials. However, engineering such materials with precisely tuned properties requires measuring how the dynamics depend on the network composition and synthesis methods. Quantifying such dynamics is challenged by variations across the time, space, and formulation space of composite networks. The protocol here describes how the Fourier analysis technique, differential dynamic microscopy (DDM), can quantify the dynamics of biopolymer networks and is particularly well suited for studies of cytoskeleton networks. DDM works on time sequences of images acquired using a range of microscopy modalities, including laser-scanning confocal, widefield fluorescence, and brightfield imaging. From such image sequences, one can extract characteristic decorrelation times of density fluctuations across a span of wave vectors. A user-friendly, open-source Python package to perform DDM analysis is also developed. With this package, one can measure the dynamics of labeled cytoskeleton components or of embedded tracer particles, as demonstrated here with data of intermediate filament (vimentin) networks and active actin-microtubule networks. Users with no prior programming or image processing experience will be able to perform DDM using this software package and associated documentation.
Collapse
Affiliation(s)
- Hannah N Verwei
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University
| | - Gloria Lee
- Department of Physics and Biophysics, University of San Diego
| | - Gregor Leech
- Department of Physics and Biophysics, University of San Diego
| | - Irene Istúriz Petitjean
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology
| | - Gijsje H Koenderink
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology
| | | | | |
Collapse
|
48
|
Salvador J, Hernandez GE, Ma F, Abrahamson CW, Pellegrini M, Goldman R, Ridge KM, Iruela-Arispe ML. Transcriptional Evaluation of the Ductus Arteriosus at the Single-Cell Level Uncovers a Requirement for Vim (Vimentin) for Complete Closure. Arterioscler Thromb Vasc Biol 2022; 42:732-742. [PMID: 35443793 PMCID: PMC9806842 DOI: 10.1161/atvbaha.121.317172] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Failure to close the ductus arteriosus, patent ductus arteriosus, accounts for 10% of all congenital heart defects. Despite significant advances in patent ductus arteriosus management, including pharmacological treatment targeting the prostaglandin pathway, a proportion of patients fail to respond and must undergo surgical intervention. Thus, further refinement of the cellular and molecular mechanisms that govern vascular remodeling of this vessel is required. METHODS We performed single-cell RNA-sequencing of the ductus arteriosus in mouse embryos at E18.5 (embryonic day 18.5), and P0.5 (postnatal day 0.5), and P5 to identify transcriptional alterations that might be associated with remodeling. We further confirmed our findings using transgenic mouse models coupled with immunohistochemistry analysis. RESULTS The intermediate filament vimentin emerged as a candidate that might contribute to closure of the ductus arteriosus. Indeed, mice with genetic deletion of vimentin fail to complete vascular remodeling of the ductus arteriosus. To seek mechanisms, we turned to the RNA-sequencing data that indicated changes in Jagged1 with similar profile to vimentin and pointed to potential links with Notch. In fact, Notch3 signaling was impaired in vimentin null mice and vimentin null mice phenocopies patent ductus arteriosus in Jagged1 endothelial and smooth muscle deleted mice. CONCLUSIONS Through single-cell RNA-sequencing and by tracking closure of the ductus arteriosus in mice, we uncovered the unexpected contribution of vimentin in driving complete closure of the ductus arteriosus through a mechanism that includes deregulation of the Notch signaling pathway.
Collapse
Affiliation(s)
- Jocelynda Salvador
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago
| | - Gloria E Hernandez
- Molecular Biology Institute (G.E.H., F.M.), University of California, Los Angeles
| | - Feiyang Ma
- Molecular Biology Institute (G.E.H., F.M.), University of California, Los Angeles
| | - Cyrus W Abrahamson
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago
| | - Matteo Pellegrini
- Department of Molecular, Cell and Development Biology (M.P.), University of California, Los Angeles
| | - Robert Goldman
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago
| | - Karen M Ridge
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago.,Department of Medicine, Feinberg School of Medicine (K.M.R.), Northwestern University, Chicago
| | - M Luisa Iruela-Arispe
- Department of Cell and Development Biology (J.S., C.W.A., R.G., K.M.R., M.L.I.-A.), Northwestern University, Chicago
| |
Collapse
|
49
|
Hang JT, Xu GK, Gao H. Frequency-dependent transition in power-law rheological behavior of living cells. SCIENCE ADVANCES 2022; 8:eabn6093. [PMID: 35522746 PMCID: PMC9075802 DOI: 10.1126/sciadv.abn6093] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Living cells are active viscoelastic materials exhibiting diverse mechanical behaviors at different time scales. However, dynamical rheological characteristics of cells in frequency range spanning many orders of magnitude, especially in high frequencies, remain poorly understood. Here, we show that a self-similar hierarchical model can capture cell's power-law rheological characteristics in different frequency scales. In low-frequency scales, the storage and loss moduli exhibit a weak power-law dependence on frequency with same exponent. In high-frequency scales, the storage modulus becomes a constant, while the loss modulus shows a power-law dependence on frequency with an exponent of 1.0. The transition between low- and high-frequency scales is defined by a transition frequency based on cell's mechanical parameters. The cytoskeletal differences of different cell types or states can be characterized by changes in mechanical parameters in the model. This study provides valuable insights into potentially using mechanics-based markers for cell classification and cancer diagnosis.
Collapse
Affiliation(s)
- Jiu-Tao Hang
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, SVL, School of Aerospace Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Guang-Kui Xu
- Laboratory for Multiscale Mechanics and Medical Science, Department of Engineering Mechanics, SVL, School of Aerospace Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- Corresponding author. (G.-K.X.); (H.G.)
| | - Huajian Gao
- School of Mechanical and Aerospace Engineering, College of Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Institute of High Performance Computing, A*STAR, Singapore 138632, Singapore
- Corresponding author. (G.-K.X.); (H.G.)
| |
Collapse
|
50
|
Mary G, Mazuel F, Nier V, Fage F, Nagle I, Devaud L, Bacri JC, Asnacios S, Asnacios A, Gay C, Marcq P, Wilhelm C, Reffay M. All-in-one rheometry and nonlinear rheology of multicellular aggregates. Phys Rev E 2022; 105:054407. [PMID: 35706238 DOI: 10.1103/physreve.105.054407] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 03/24/2022] [Indexed: 06/15/2023]
Abstract
Tissues are generally subjected to external stresses, a potential stimulus for their differentiation or remodeling. While single-cell rheology has been extensively studied leading to controversial results about nonlinear response, mechanical tissue behavior under external stress is still poorly understood, in particular, the way individual cell properties translate at the tissue level. Herein, using magnetic cells we were able to form perfectly monitored cellular aggregates (magnetic molding) and to deform them under controlled applied stresses over a wide range of timescales and amplitudes (magnetic rheometer). We explore the rheology of these minimal tissue models using both standard assays (creep and oscillatory response) as well as an innovative broad spectrum solicitation coupled with inference analysis thus being able to determine in a single experiment the best rheological model. We find that multicellular aggregates exhibit a power-law response with nonlinearities leading to tissue stiffening at high stress. Moreover, we reveal the contribution of intracellular (actin network) and intercellular components (cell-cell adhesions) in this aggregate rheology.
Collapse
Affiliation(s)
- Gaëtan Mary
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| | - François Mazuel
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| | - Vincent Nier
- Laboratoire Physico Chimie Curie, UMR 168, CNRS, Institut Curie, PSL University, Sorbonne Université, 75005 Paris, France
| | - Florian Fage
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| | - Irène Nagle
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| | - Louisiane Devaud
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| | - Jean-Claude Bacri
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| | - Sophie Asnacios
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
- Faculty of Science and Engineering, UFR 925 Physics, Sorbonne Université, Paris France
| | - Atef Asnacios
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| | - Cyprien Gay
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| | - Philippe Marcq
- Laboratoire Physico Chimie Curie, UMR 168, CNRS, Institut Curie, PSL University, Sorbonne Université, 75005 Paris, France
- Faculty of Science and Engineering, UFR 925 Physics, Sorbonne Université, Paris France
- Laboratoire Physique et Mécanique des Matériaux Hétérogènes, CNRS, ESPCI Paris, PSL University, Sorbonne Université, Université de Paris Cité, 75005 Paris, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
- Laboratoire Physico Chimie Curie, UMR 168, CNRS, Institut Curie, PSL University, Sorbonne Université, 75005 Paris, France
| | - Myriam Reffay
- Laboratoire Matière et Systèmes Complexes, UMR 7057, CNRS and Université de Paris Cité, 75205 Paris cedex 13, France
| |
Collapse
|