1
|
Lima I, Silva A, Sousa F, Ferreira W, Freire R, de Oliveira C, de Sousa J. Measuring the viscoelastic relaxation function of cells with a time-dependent interpretation of the Hertz-Sneddon indentation model. Heliyon 2024; 10:e30623. [PMID: 38770291 PMCID: PMC11103437 DOI: 10.1016/j.heliyon.2024.e30623] [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: 02/06/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 05/22/2024] Open
Abstract
The Hertz-Sneddon elastic indentation model is widely adopted in the biomechanical investigation of living cells and other soft materials using atomic force microscopy despite the explicit viscoelastic nature of these materials. In this work, we demonstrate that an exact analytical viscoelastic force model for power-law materials, can be interpreted as a time-dependent Hertz-Sneddon-like model. Characterizing fibroblasts (L929) and osteoblasts (OFCOLII) demonstrates the model's accuracy. Our results show that the difference between Young's modulus E Y obtained by fitting force curves with the Hertz-Sneddon model and the effective Young's modulus derived from the viscoelastic force model is less than 3%, even when cells are probed at large forces where nonlinear deformation effects become significant. We also propose a measurement protocol that involves probing samples at different indentation speeds and forces, enabling the construction of the average viscoelastic relaxation function of samples by conveniently fitting the force curves with the Hertz-Sneddon model.
Collapse
Affiliation(s)
- I.V.M. Lima
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - A.V.S. Silva
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
- Instituto Federal do Rio Grande do Norte, Pau dos Ferros, 59900-000, Rio Grande do Norte, Brazil
| | - F.D. Sousa
- Núcleo de Biologia Experimental, Universidade de Fortaleza, Fortaleza, 60811-905, Ceará, Brazil
| | - W.P. Ferreira
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - R.S. Freire
- Central Analítica, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - C.L.N. de Oliveira
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| | - J.S. de Sousa
- Departamento de Física, Universidade Federal do Ceará, Fortaleza, 60440-900, Ceará, Brazil
| |
Collapse
|
2
|
Massey A, Stewart J, Smith C, Parvini C, McCormick M, Do K, Cartagena-Rivera AX. Mechanical properties of human tumour tissues and their implications for cancer development. NATURE REVIEWS. PHYSICS 2024; 6:269-282. [PMID: 38706694 PMCID: PMC11066734 DOI: 10.1038/s42254-024-00707-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 05/07/2024]
Abstract
The mechanical properties of cells and tissues help determine their architecture, composition and function. Alterations to these properties are associated with many diseases, including cancer. Tensional, compressive, adhesive, elastic and viscous properties of individual cells and multicellular tissues are mostly regulated by reorganization of the actomyosin and microtubule cytoskeletons and extracellular glycocalyx, which in turn drive many pathophysiological processes, including cancer progression. This Review provides an in-depth collection of quantitative data on diverse mechanical properties of living human cancer cells and tissues. Additionally, the implications of mechanical property changes for cancer development are discussed. An increased knowledge of the mechanical properties of the tumour microenvironment, as collected using biomechanical approaches capable of multi-timescale and multiparametric analyses, will provide a better understanding of the complex mechanical determinants of cancer organization and progression. This information can lead to a further understanding of resistance mechanisms to chemotherapies and immunotherapies and the metastatic cascade.
Collapse
Affiliation(s)
- Andrew Massey
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Jamie Stewart
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Chynna Smith
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
- These authors contributed equally: Jamie Stewart, Chynna Smith
| | - Cameron Parvini
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Moira McCormick
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Kun Do
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Alexander X. Cartagena-Rivera
- Section on Mechanobiology, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
3
|
Cambria E, Coughlin MF, Floryan MA, Offeddu GS, Shelton SE, Kamm RD. Linking cell mechanical memory and cancer metastasis. Nat Rev Cancer 2024; 24:216-228. [PMID: 38238471 PMCID: PMC11146605 DOI: 10.1038/s41568-023-00656-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/07/2023] [Indexed: 03/01/2024]
Abstract
Metastasis causes most cancer-related deaths; however, the efficacy of anti-metastatic drugs is limited by incomplete understanding of the biological mechanisms that drive metastasis. Focusing on the mechanics of metastasis, we propose that the ability of tumour cells to survive the metastatic process is enhanced by mechanical stresses in the primary tumour microenvironment that select for well-adapted cells. In this Perspective, we suggest that biophysical adaptations favourable for metastasis are retained via mechanical memory, such that the extent of memory is influenced by both the magnitude and duration of the mechanical stress. Among the mechanical cues present in the primary tumour microenvironment, we focus on high matrix stiffness to illustrate how it alters tumour cell proliferation, survival, secretion of molecular factors, force generation, deformability, migration and invasion. We particularly centre our discussion on potential mechanisms of mechanical memory formation and retention via mechanotransduction and persistent epigenetic changes. Indeed, we propose that the biophysical adaptations that are induced by this process are retained throughout the metastatic process to improve tumour cell extravasation, survival and colonization in the distant organ. Deciphering mechanical memory mechanisms will be key to discovering a new class of anti-metastatic drugs.
Collapse
Affiliation(s)
- Elena Cambria
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Mark F Coughlin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marie A Floryan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Giovanni S Offeddu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sarah E Shelton
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
4
|
do Nascimento Amorim MDS, Silva França ÁR, Santos-Oliveira R, Rodrigues Sanches J, Marinho Melo T, Araújo Serra Pinto B, Barbosa LRS, Alencar LMR. Atomic Force Microscopy Applied to the Study of Tauopathies. ACS Chem Neurosci 2024; 15:699-715. [PMID: 38305187 DOI: 10.1021/acschemneuro.3c00819] [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: 02/03/2024] Open
Abstract
Atomic force microscopy (AFM) is a scanning probe microscopy technique which has a physical principle, the measurement of interatomic forces between a very thin tip and the surface of a sample, allowing the obtaining of quantitative data at the nanoscale, contributing to the surface study and mechanical characterization. Due to its great versatility, AFM has been used to investigate the structural and nanomechanical properties of several inorganic and biological materials, including neurons affected by tauopathies. Tauopathies are neurodegenerative diseases featured by aggregation of phosphorylated tau protein inside neurons, leading to functional loss and progressive neurotoxicity. In the broad universe of neurodegenerative diseases, tauopathies comprise the most prevalent, with Alzheimer's disease as its main representative. This review highlights the use of AFM as a suitable research technique for the study of cellular damages in tauopathies, even in early stages, allowing elucidation of pathogenic mechanisms of these diseases.
Collapse
Affiliation(s)
- Maria do Socorro do Nascimento Amorim
- Laboratory of Biophysics and Nanosystems, Department of Physics, Federal University of Maranhão, Campus Bacanga, São Luís 65080-805, Maranhão, Brazil
| | - Álefe Roger Silva França
- Laboratory of Biophysics and Nanosystems, Department of Physics, Federal University of Maranhão, Campus Bacanga, São Luís 65080-805, Maranhão, Brazil
| | - Ralph Santos-Oliveira
- Nuclear Engineering Institute, Brazilian Nuclear Energy Commission, Rio de Janeiro 21941906, Brazil
- Laboratory of Nanoradiopharmacy, Rio de Janeiro State University, Rio de Janeiro 23070200, Brazil
| | - Jonas Rodrigues Sanches
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, Campus Bacanga, São Luís, 65080-805, Maranhão, Brazil
| | - Thamys Marinho Melo
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, Campus Bacanga, São Luís, 65080-805, Maranhão, Brazil
| | - Bruno Araújo Serra Pinto
- Laboratory of Experimental Physiology, Department of Physiological Sciences, Federal University of Maranhão, Campus Bacanga, São Luís, 65080-805, Maranhão, Brazil
| | - Leandro R S Barbosa
- Department of General Physics, Institute of Physics, University of São Paulo, São Paulo 05508-000, SP, Brazil
- Brazilian Synchrotron Light Laboratory (LNLS), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, SP, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Laboratory of Biophysics and Nanosystems, Department of Physics, Federal University of Maranhão, Campus Bacanga, São Luís 65080-805, Maranhão, Brazil
| |
Collapse
|
5
|
Tulchinsky M, Weihs D. Mechanobiological cell adaptations to changing microenvironments determine cancer invasiveness: Experimentally validated finite element modeling. J Biomed Mater Res A 2023; 111:1951-1959. [PMID: 37606496 DOI: 10.1002/jbm.a.37597] [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: 04/17/2023] [Revised: 07/30/2023] [Accepted: 08/11/2023] [Indexed: 08/23/2023]
Abstract
Metastases are the leading cause of cancer-associated deaths. A key process in metastasis is cell invasiveness, which is driven and controlled by cancer cell interactions with their microenvironment. We have previously shown that invasive cancer cells forcefully push into and indent physiological stiffness gels to cell-scale depths, where the percentage of indenting cells and their attained depths provide clinically relevant predictions of tumor invasiveness and the potential metastatic risk. The cell-attained, invasive indentation depths are directly affected by gel-microenvironment mechanics, which can concurrently modulate the cells' mechanics and force application capacity, in a complex, coordinated mechanobiological response. As it is impossible to experimentally isolate the different contributions of cell and gel mechanics to cancer cell invasiveness, we perform finite element modeling with literature-based parameters. Under average-scale, cell cytoplasm and nucleus mechanics and cell-applied force levels, increasing gel stiffness 1-50 kPa significantly reduced the attained indentation depth by >200%, while the gel's Poisson ratio reduced depths only by up to 20% and only when the ratio was >0.4; this reveals microenvironment mechanics that can promote invasiveness. Experiments with varying-invasiveness cancer cells exhibited qualitative variations in their responses to gel stiffness increase, for example large/small reduction in indentation depth or increase and then reduction. We quantitatively and qualitatively reproduced the different experimental responses via coordinated changes in cell mechanics and applied force levels. Thus, the different cancer cell capacities to adapt their mechanobiology in response to mechanically changing microenvironments likely determine the varying cancer invasiveness and metastatic risk levels in patients.
Collapse
Affiliation(s)
- Marina Tulchinsky
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Daphne Weihs
- Faculty of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
6
|
Gilbert RJC. Peptide-based pore formation and cell membrane deformation: European Biophysics Journal Prizes at EBSA 2023. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2023; 52:619-623. [PMID: 37994943 DOI: 10.1007/s00249-023-01691-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
The European Biophysics Journal Prizes awarded at the European Biophysical Societies Association (EBSA) Congress in Stockholm in the Summer of 2023 recognised papers published in 2020 and 2021 which made use of multiple complementing experimental, theoretical and computational approaches. One of the winning papers addressed the specific role of arginine residues within antimicrobial and cell-penetrating peptides, in promoting membrane defect stabilisation and pore formation. The other winning paper described the influence of atomic force microscopy probe geometry on the measurement of surface deformability, assessed for investigation of the differing viscoelastic properties of non-malignant and cancerous cells. These papers showcase biophysical science; the importance of combining different experimental, modelling and molecular dynamics methods; and how researchers need to understand the theoretical basis and the limitations of the techniques they use. EBSA warmly congratulates the authors on their work and its subsequent recognition. Publication of these papers also demonstrates the ongoing commitment of the European Biophysics Journal to molecular scale and to systems biophysics, and to support of the international biophysical community.
Collapse
Affiliation(s)
- Robert J C Gilbert
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK.
| |
Collapse
|
7
|
Pérez-Domínguez S, Kulkarni SG, Pabijan J, Gnanachandran K, Holuigue H, Eroles M, Lorenc E, Berardi M, Antonovaite N, Marini ML, Lopez Alonso J, Redonto-Morata L, Dupres V, Janel S, Acharya S, Otero J, Navajas D, Bielawski K, Schillers H, Lafont F, Rico F, Podestà A, Radmacher M, Lekka M. Reliable, standardized measurements for cell mechanical properties. NANOSCALE 2023; 15:16371-16380. [PMID: 37789717 DOI: 10.1039/d3nr02034g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Atomic force microscopy (AFM) has become indispensable for studying biological and medical samples. More than two decades of experiments have revealed that cancer cells are softer than healthy cells (for measured cells cultured on stiff substrates). The softness or, more precisely, the larger deformability of cancer cells, primarily independent of cancer types, could be used as a sensitive marker of pathological changes. The wide application of biomechanics in clinics would require designing instruments with specific calibration, data collection, and analysis procedures. For these reasons, such development is, at present, still very limited, hampering the clinical exploitation of mechanical measurements. Here, we propose a standardized operational protocol (SOP), developed within the EU ITN network Phys2BioMed, which allows the detection of the biomechanical properties of living cancer cells regardless of the nanoindentation instruments used (AFMs and other indenters) and the laboratory involved in the research. We standardized the cell cultures, AFM calibration, measurements, and data analysis. This effort resulted in a step-by-step SOP for cell cultures, instrument calibration, measurements, and data analysis, leading to the concordance of the results (Young's modulus) measured among the six EU laboratories involved. Our results highlight the importance of the SOP in obtaining a reproducible mechanical characterization of cancer cells and paving the way toward exploiting biomechanics for diagnostic purposes in clinics.
Collapse
Affiliation(s)
| | - Shruti G Kulkarni
- Institute of Biophysics, University of Bremen, 28359, Bremen, Germany.
| | - Joanna Pabijan
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Kraków, Poland.
| | - Kajangi Gnanachandran
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Kraków, Poland.
| | - Hatice Holuigue
- Department of Physics "Aldo Pontremoli" and CIMAINA, University of Milano, via Celoria 16, 20133 Milano, Italy.
| | - Mar Eroles
- Aix-Marseille Univ., CNRS, INSERM, LAI, Turing Centre for Living Systems, Marseille, France
| | - Ewelina Lorenc
- Department of Physics "Aldo Pontremoli" and CIMAINA, University of Milano, via Celoria 16, 20133 Milano, Italy.
| | - Massimiliano Berardi
- Laserlab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, De Boelelaan 1081, 1081 HV, Amsterdam, The Netherlands
- Optics11 Life, Hettenheuvelweg 37-39, 1101 BM, Amsterdam, The Netherlands
| | - Nelda Antonovaite
- Optics11 Life, Hettenheuvelweg 37-39, 1101 BM, Amsterdam, The Netherlands
| | - Maria Luisa Marini
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Javier Lopez Alonso
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Lorena Redonto-Morata
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Vincent Dupres
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Sebastien Janel
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Sovon Acharya
- Institute of Physiology II, University Muenster, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Jorge Otero
- Institute for Bioengineering of Catalonia and Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Daniel Navajas
- Institute for Bioengineering of Catalonia and Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Kevin Bielawski
- Optics11 Life, Hettenheuvelweg 37-39, 1101 BM, Amsterdam, The Netherlands
| | - Hermann Schillers
- Institute of Physiology II, University Muenster, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Frank Lafont
- Université de Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR9017, CIIL-Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Felix Rico
- Aix-Marseille Univ., CNRS, INSERM, LAI, Turing Centre for Living Systems, Marseille, France
| | - Alessandro Podestà
- Department of Physics "Aldo Pontremoli" and CIMAINA, University of Milano, via Celoria 16, 20133 Milano, Italy.
| | - Manfred Radmacher
- Institute of Biophysics, University of Bremen, 28359, Bremen, Germany.
| | - Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Kraków, Poland.
| |
Collapse
|
8
|
Daniel C, Traub F, Sachsenmaier S, Riester R, Mederake M, Konrads C, Danalache M. An exploratory study of cell stiffness as a mechanical label-free biomarker across multiple musculoskeletal sarcoma cells. BMC Cancer 2023; 23:862. [PMID: 37700272 PMCID: PMC10498616 DOI: 10.1186/s12885-023-11375-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Cancer cells are characterized by changes in cell cytoskeletal architecture and stiffness. Despite advances in understanding the molecular mechanisms of musculoskeletal cancers, the corresponding cellular mechanical properties remain largely unexplored. The aim of this study was to investigate the changes in cellular stiffness and the associated cytoskeleton configuration alterations in various musculoskeletal cancer cells. METHODS Cell lines from five main sarcoma types of the musculoskeletal system (chondrosarcoma, osteosarcoma, Ewing sarcoma, fibrosarcoma and rhabdomyosarcoma) as well as their healthy cell counterparts (chondrocytes, osteoblasts, mesenchymal stem cells, fibroblasts, skeletal muscle cells) were subjected to cell stiffness measurements via atomic force microscopy (AFM). Biochemical and structural changes of the cytoskeleton (F-actin, β-tubulin and actin-related protein 2/3) were assessed by means of fluorescence labelling, ELISA and qPCR. RESULTS While AFM stiffness measurements showed that the majority of cancer cells (osteosarcoma, Ewing sarcoma, fibrosarcoma and rhabdomyosarcoma) were significantly less stiff than their corresponding non-malignant counterparts (p < 0.001), the chondrosarcoma cells were significant stiffer than the chondrocytes (p < 0.001). Microscopically, the distribution of F-actin differed between malignant entities and healthy counterparts: the organisation in well aligned stress fibers was disrupted in cancer cell lines and the proteins was mainly concentrated at the periphery of the cell, whereas β-tubulin had a predominantly perinuclear localization. While the F-actin content was lower in cancer cells, particularly Ewing sarcoma (p = 0.018) and Fibrosarcoma (p = 0.023), this effect was even more pronounced in the case of β-tubulin for all cancer-healthy cell duos. Interestingly, chondrosarcoma cells were characterized by a significant upregulation of β-tubulin gene expression (p = 0.005) and protein amount (p = 0.032). CONCLUSION Modifications in cellular stiffness, along with structural and compositional cytoskeleton rearrangement, constitute typical features of sarcomas cells, when compared to their healthy counterpart. Notably, whereas a decrease in stiffness is typically a feature of malignant entities, chondrosarcoma cells were stiffer than chondrocytes, with chondrosarcoma cells exhibiting a significantly upregulated β-tubulin expression. Each Sarcoma entity may have his own cellular-stiffness and cytoskeleton organisation/composition fingerprint, which in turn may be exploited for diagnostic or therapeutic purposes.
Collapse
Affiliation(s)
- Cyril Daniel
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany.
- Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany.
| | - Frank Traub
- Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
- Department of Orthopedics and Traumatology, University Medical Center Mainz, Johannes Gutenberg-University Mainz, 55122, Mainz, Germany
| | - Saskia Sachsenmaier
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
- Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
| | - Rosa Riester
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
| | - Moritz Mederake
- Department of Trauma and Reconstructive Surgery, BG Clinic, University of Tübingen, 72076, Tübingen, Germany
| | - Christian Konrads
- Department of Orthopedics and Traumatology, Hanseatic Hospital Stralsund, 18437, Stralsund, Germany
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
- Department of Orthopedic Surgery, University Hospital of Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
9
|
Han SJ, Kwon S, Kim KS. Contribution of mechanical homeostasis to epithelial-mesenchymal transition. Cell Oncol (Dordr) 2022; 45:1119-1136. [PMID: 36149601 DOI: 10.1007/s13402-022-00720-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Metastasis refers to the spread of cancer cells from a primary tumor to other parts of the body via the lymphatic system and bloodstream. With tremendous effort over the past decades, remarkable progress has been made in understanding the molecular and cellular basis of metastatic processes. Metastasis occurs through five steps, including infiltration and migration, intravasation, survival, extravasation, and colonization. Various molecular and cellular factors involved in the metastatic process have been identified, such as epigenetic factors of the extracellular matrix (ECM), cell-cell interactions, soluble signaling, adhesion molecules, and mechanical stimuli. However, the underlying cause of cancer metastasis has not been elucidated. CONCLUSION In this review, we have focused on changes in the mechanical properties of cancer cells and their surrounding environment to understand the causes of cancer metastasis. Cancer cells have unique mechanical properties that distinguish them from healthy cells. ECM stiffness is involved in cancer cell growth, particularly in promoting the epithelial-mesenchymal transition (EMT). During tumorigenesis, the mechanical properties of cancer cells change in the direction opposite to their environment, resulting in a mechanical stress imbalance between the intracellular and extracellular domains. Disruption of mechanical homeostasis may be one of the causes of EMT that triggers the metastasis of cancer cells.
Collapse
Affiliation(s)
- Se Jik Han
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, Seoul, Korea.,Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea
| | - Sangwoo Kwon
- Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea
| | - Kyung Sook Kim
- Department of Biomedical Engineering, Graduate School, Kyung Hee University, Seoul, Korea.
| |
Collapse
|
10
|
Zamora-Ceballos M, Bárcena J, Mertens J. Eukaryotic CRFK Cells Motion Characterized with Atomic Force Microscopy. Int J Mol Sci 2022; 23:ijms232214369. [PMID: 36430849 PMCID: PMC9692694 DOI: 10.3390/ijms232214369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
We performed a time-lapse imaging with atomic force microscopy (AFM) of the motion of eukaryotic CRFK (Crandell-Rees Feline Kidney) cells adhered onto a glass surface and anchored to other cells in culture medium at 37 °C. The main finding is a gradient in the spring constant of the actomyosin cortex along the cells axis. The rigidity increases at the rear of the cells during motion. This observation as well as a dramatic decrease of the volume suggests that cells may organize a dissymmetry in the skeleton network to expulse water and drive actively the rear edge.
Collapse
Affiliation(s)
- María Zamora-Ceballos
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Valdeolmos, 28130 Madrid, Spain
| | - Juan Bárcena
- Centro de Investigación en Sanidad Animal (CISA-INIA/CSIC), Valdeolmos, 28130 Madrid, Spain
| | - Johann Mertens
- Madrid Institute for Advanced Studies in Nanoscience (IMDEA Nanoscience), Campus de Cantoblanco, 28049 Madrid, Spain
- Correspondence:
| |
Collapse
|
11
|
Bory Prevez H, Soutelo Jimenez AA, Roca Oria EJ, Heredia Kindelán JA, Morales González M, Villar Goris NA, Hernández Mesa N, Sierra González VG, Infantes Frometa Y, Montijano JI, Cabrales LEB. Simulations of surface charge density changes during the untreated solid tumour growth. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220552. [PMID: 36465673 PMCID: PMC9709566 DOI: 10.1098/rsos.220552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/10/2022] [Indexed: 06/17/2023]
Abstract
Understanding untreated tumour growth kinetics and its intrinsic behaviour is interesting and intriguing. The aim of this study is to propose an approximate analytical expression that allows us to simulate changes in surface charge density at the cancer-surrounding healthy tissue interface during the untreated solid tumour growth. For this, the Gompertz and Poisson equations are used. Simulations reveal that the unperturbed solid tumour growth is closely related to changes in the surface charge density over time between the tumour and the surrounding healthy tissue. Furthermore, the unperturbed solid tumour growth is governed by temporal changes in this surface charge density. It is concluded that results corroborate the correspondence between the electrical and physiological parameters in the untreated cancer, which may have an essential role in its growth, progression, metastasis and protection against immune system attack and anti-cancer therapies. In addition, the knowledge of surface charge density changes at the cancer-surrounding healthy tissue interface may be relevant when redesigning the molecules in chemotherapy and immunotherapy taking into account their polarities. This can also be true in the design of completely novel therapies.
Collapse
Affiliation(s)
- Henry Bory Prevez
- Departamento de Control Automático, Facultad de Ingeniería Eléctrica, Universidad de Oriente, Santiago de Cuba, Cuba
| | | | - Eduardo José Roca Oria
- Departamento de Física, Facultad de Ciencias Naturales y Exactas, Universidad de Oriente, Santiago de Cuba, Cuba
| | | | - Maraelys Morales González
- Departamento de Farmacia, Facultad de Ciencias Naturales y Exactas, Universidad de Oriente, Santiago de Cuba, Cuba
| | - Narciso Antonio Villar Goris
- Departamento de Ciencia e Innovación, Centro Nacional de Electromagnetismo Aplicado, Universidad de Oriente, Santiago de Cuba, Cuba
- Universidad Autónoma de Santo Domingo, Santo Domingo, República Dominicana
| | | | | | | | - Juan Ignacio Montijano
- Departamento de Matemática Aplicada, Instituto Universitario de Matemática y Aplicaciones, Universidad de Zaragoza, Zaragoza, España
| | - Luis Enrique Bergues Cabrales
- Departamento de Ciencia e Innovación, Centro Nacional de Electromagnetismo Aplicado, Universidad de Oriente, Santiago de Cuba, Cuba
- Departamento de Matemática Aplicada, Instituto Universitario de Matemática y Aplicaciones, Universidad de Zaragoza, Zaragoza, España
| |
Collapse
|
12
|
Vasudevan J, Jiang K, Fernandez J, Lim CT. Extracellular matrix mechanobiology in cancer cell migration. Acta Biomater 2022; 163:351-364. [PMID: 36243367 DOI: 10.1016/j.actbio.2022.10.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/11/2022] [Accepted: 10/06/2022] [Indexed: 11/01/2022]
Abstract
The extracellular matrix (ECM) is pivotal in modulating tumor progression. Besides chemically stimulating tumor cells, it also offers physical support that orchestrates the sequence of events in the metastatic cascade upon dynamically modulating cell mechanosensation. Understanding this translation between matrix biophysical cues and intracellular signaling has led to rapid growth in the interdisciplinary field of cancer mechanobiology in the last decade. Substantial efforts have been made to develop novel in vitro tumor mimicking platforms to visualize and quantify the mechanical forces within the tissue that dictate tumor cell invasion and metastatic growth. This review highlights recent findings on tumor matrix biophysical cues such as fibrillar arrangement, crosslinking density, confinement, rigidity, topography, and non-linear mechanics and their implications on tumor cell behavior. We also emphasize how perturbations in these cues alter cellular mechanisms of mechanotransduction, consequently enhancing malignancy. Finally, we elucidate engineering techniques to individually emulate the mechanical properties of tumors that could help serve as toolkits for developing and testing ECM-targeted therapeutics on novel bioengineered tumor platforms. STATEMENT OF SIGNIFICANCE: Disrupted ECM mechanics is a driving force for transitioning incipient cells to life-threatening malignant variants. Understanding these ECM changes can be crucial as they may aid in developing several efficacious drugs that not only focus on inducing cytotoxic effects but also target specific matrix mechanical cues that support and enhance tumor invasiveness. Designing and implementing an optimal tumor mimic can allow us to predictively map biophysical cue-modulated cell behaviors and facilitate the design of improved lab-grown tumor models with accurately controlled structural features. This review focuses on the abnormal changes within the ECM during tumorigenesis and its implications on tumor cell-matrix mechanoreciprocity. Additionally, it accentuates engineering approaches to produce ECM features of varying levels of complexity which is critical for improving the efficiency of current engineered tumor tissue models.
Collapse
|
13
|
Lekka M. Applicability of atomic force microscopy to determine cancer-related changes in cells. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2022; 380:20210346. [PMID: 35909354 DOI: 10.1098/rsta.2021.0346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/24/2022] [Indexed: 06/15/2023]
Abstract
The determination of mechanical properties of living cells as an indicator of cancer progression has become possible with the development of local measurement techniques such as atomic force microscopy (AFM). Its most important advantage is a nanoscopic character, implying that very local alterations can be quantified. The results gathered from AFM measurements of various cancers show that, for most cancers, individual cells are characterized by the lower apparent Young's modulus, denoting higher cell deformability. The measured value depends on various factors, like the properties of substrates used for cell growth, force loading rate or indentation depth. Despite this, the results proved the AFM capability to recognize mechanically altered cells. This can significantly impact the development of methodological approaches toward the precise identification of pathological cells. This article is part of the theme issue 'Nanocracks in nature and industry'.
Collapse
Affiliation(s)
- Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland
| |
Collapse
|
14
|
Tang X, Zhang Y, Mao J, Wang Y, Zhang Z, Wang Z, Yang H. Effects of substrate stiffness on the viscoelasticity and migration of prostate cancer cells examined by atomic force microscopy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2022; 13:560-569. [PMID: 35860456 PMCID: PMC9263554 DOI: 10.3762/bjnano.13.47] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/03/2022] [Indexed: 06/07/2023]
Abstract
The stiffness of the extracellular matrix of tumour cells plays a key role in tumour cell metastasis. However, it is unclear how mechanical properties regulate the cellular response to the environmental matrix. In this study, atomic force microscopy (AFM) and laser confocal imaging were used to qualitatively evaluate the relationship between substrate stiffness and migration of prostate cancer (PCa) cells. Cells cultured on stiff substrates (35 kPa) undergone several interesting phenomena compared to those on soft substrates (3 kPa). Here, the stimulation generated by the stiff substrates triggered the F-actin skeleton to bundle its filaments, increasing the polarity index of the external contour of PCa cells. Analysis of AFM force-distance curves indicated that the elasticity of the cells cultured on 35 kPa substrates increased while the viscosity decreased. Wound-healing experiments showed that PCa cells cultured on 35 kPa substrates have higher migration potential. These phenomena suggested that the mechanical properties may be correlated with the migration of PCa cells. After actin depolymerisation, the elasticity of the PCa cells decreased while the viscosity increased, and the migration ability was correspondingly decreased. In conclusion, this study clearly demonstrated the relationship between substrate stiffness and the mechanical properties of cells in prostate tumour metastasis, providing a basis for understanding the changes in the biomechanical properties at a single-cell level.
Collapse
Affiliation(s)
- Xiaoqiong Tang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Yan Zhang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Jiangbing Mao
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Yuhua Wang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Zhenghong Zhang
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Zhengchao Wang
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350117, China
| | - Hongqin Yang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| |
Collapse
|
15
|
Hang X, He S, Dong Z, Li Y, Huang Z, Zhang Y, Sun H, Lin L, Li H, Wang Y, Liu B, Wu N, Ren T, Fan Y, Lou J, Yang R, Jiang L, Chang L. High-Throughput DNA Tensioner Platform for Interrogating Mechanical Heterogeneity of Single Living Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106196. [PMID: 35322558 DOI: 10.1002/smll.202106196] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/24/2021] [Indexed: 06/14/2023]
Abstract
Cell mechanical forces play fundamental roles in regulating cellular responses to environmental stimulations. The shortcomings of conventional methods, including force resolution and cellular throughput, make them less accessible to mechanical heterogeneity at the single-cell level. Here, a DNA tensioner platform is introduced with high throughput (>10 000 cells per chip) and pN-level resolution. A microfluidic-based cell array is trapped on "hairpin-structured" DNA tensioners that enable transformation of the mechanical information of living cells into fluorescence signals. By using the platform, one can identify enhanced mechanical forces of drug-resistant cells as compared to their drug-sensitive counterparts, and mechanical differences between metastatic tumor cells in pleural effusion and nonmetastatic histiocytes. Further genetic analysis traces two genes, VEGFA and MINK1, that may play deterministic roles in regulating mechanical heterogeneities. In view of the ubiquity of cells' mechanical forces in the extracellular microenvironment (ECM), this platform shows wide potential to establish links of cellular mechanical heterogeneity to genetic heterogeneity.
Collapse
Affiliation(s)
- Xinxin Hang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Shiqi He
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Zaizai Dong
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Yun Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Zheng Huang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
| | - Yanruo Zhang
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, No. 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Hong Sun
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Long Lin
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Hu Li
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Yang Wang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Bing Liu
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Nan Wu
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, No. 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Tianling Ren
- Beijing National Research Center for Information Science and Technology (BNRist), Institute of Microelectronics, Tsinghua University, No. 30 Shuangqing Road, Haidian District, Beijing, 100084, China
| | - Yubo Fan
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
| | - Jizhong Lou
- Key Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, No. 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Ruiguo Yang
- Nebraska Center for Integrated Biomolecular Communication, Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Lan Jiang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, and China National Center for Bioinformation, Chinese Academy of Sciences, No. 1 Beichen West Road, Chaoyang District, Beijing, 100101, China
- College of Future Technology, and Sino-Danish College, University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing, 100049, China
| | - Lingqian Chang
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, No. 37 Xueyuan Road, Haidian District, Beijing, 100083, China
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, China
| |
Collapse
|
16
|
Huang K, Liu J, Chen Q, Feng D, Wu H, Aldanakh A, Jian Y, Xu Z, Wang S, Yang D. The effect of mechanical force in genitourinary malignancies. Expert Rev Anticancer Ther 2021; 22:53-64. [PMID: 34726963 DOI: 10.1080/14737140.2022.2000864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Mechanical force is attributed to the formation of tumor blood vessels, influences cancer cell invasion and metastasis, and promotes reprogramming of the energy metabolism. Currently, therapy strategies for the tumor microenvironment are being developed progressively. The purpose of this article is to discuss the molecular mechanism, diagnosis, and treatment of mechanical force in urinary tract cancers and outline the medications used in the mechanical microenvironment. AREAS COVERED This review covers the complex mechanical elements in the microenvironment of urinary system malignancies, focusing on mechanical molecular mechanisms for diagnosis and treatment. EXPERT OPINION The classification of various mechanical forces, such as matrix stiffness, shear force, and other forces, is relatively straightforward. However, little is known about the molecular process of mechanical forces in urinary tract malignancies. Because mechanical therapy is still controversial, it is critical to understand the molecular basis of mechanical force before adding mechanical therapy solutions.
Collapse
Affiliation(s)
- Kai Huang
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Junqiang Liu
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Qiwei Chen
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China.,School of Information Science and Technology, Dalian Maritime University, Dalian City, China
| | - Dan Feng
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Haotian Wu
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Abdullah Aldanakh
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuli Jian
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Zhongyang Xu
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Shujing Wang
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Deyong Yang
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
17
|
Labat B, Buchbinder N, Morin-Grognet S, Ladam G, Atmani H, Vannier JP. Biomimetic matrix for the study of neuroblastoma cells: A promising combination of stiffness and retinoic acid. Acta Biomater 2021; 135:383-392. [PMID: 34407473 DOI: 10.1016/j.actbio.2021.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023]
Abstract
Neuroblastoma is the third most common pediatric cancer composed of malignant immature cells that are usually treated pharmacologically by all trans-retinoic acid (ATRA) but sometimes, they can spontaneously differentiate into benign forms. In that context, biomimetic cell culture models are warranted tools as they can recapitulate many of the biochemical and biophysical cues of normal or pathological microenvironments. Inspired by that challenge, we developed a neuroblastoma culture system based on biomimetic LbL films of physiological biochemical composition and mechanical properties. For that, we used chondroitin sulfate A (CSA) and poly-L-lysine (PLL) that were assembled and mechanically tuned by crosslinking with genipin (GnP), a natural biocompatible crosslinker, in a relevant range of stiffness (30-160 kPa). We then assessed the adhesion, survival, motility, and differentiation of LAN-1 neuroblastoma cells. Remarkably, increasing the stiffness of the LbL films induced neuritogenesis that was strengthened by the combination with ATRA. These results highlight the crucial role of the mechanical cues of the neuroblastoma microenvironment since it can dramatically modulate the effect of pharmacologic drugs. In conclusion, our biomimetic platform offers a promising tool to help fundamental understanding and pharmacological screening of neuroblastoma differentiation and may assist the design of translational biomaterials to support neuronal regeneration. STATEMENT OF SIGNIFICANCE: Neuroblastoma is one of the most common pediatric tumor commonly treated by the administration of all-trans-retinoic acid (ATRA). Unfortunately, advanced neuroblastoma often develop ATRA resistance. Accordingly, in the field of pharmacological investigations on neuroblastoma, there is a tremendous need of physiologically relevant cell culture systems that can mimic normal or pathological extracellular matrices. In that context, we developed a promising matrix-like cell culture model that provides new insights on the crucial role of mechanical properties of the microenvironment upon the success of ATRA treatment on the neuroblastoma maturation. We were able to control adhesion, survival, motility, and differentiation of neuroblastoma cells. More broadly, we believe that our system will help the design of in vitro pharmacological screening strategy.
Collapse
Affiliation(s)
- Beatrice Labat
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, 55 rue Saint-Germain, 27000 Évreux, France.
| | | | - Sandrine Morin-Grognet
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, 55 rue Saint-Germain, 27000 Évreux, France
| | - Guy Ladam
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, 55 rue Saint-Germain, 27000 Évreux, France
| | - Hassan Atmani
- Normandie Univ, UNIROUEN, INSA Rouen, CNRS, PBS UMR 6270, 55 rue Saint-Germain, 27000 Évreux, France
| | - Jean-Pierre Vannier
- Normandie Univ, UNIROUEN, PANTHER - INSERM 1234 - UFR de Médecine et de Pharmacie de Rouen 22, boulevard Gambetta 76000 Rouen, France
| |
Collapse
|
18
|
Li M, Xi N, Liu L. Hierarchical micro-/nanotopography for tuning structures and mechanics of cells probed by atomic force microscopy. IEEE Trans Nanobioscience 2021; 20:543-553. [PMID: 34242170 DOI: 10.1109/tnb.2021.3096056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Extracellular matrix plays an important role in regulating the behaviors of cells, and utilizing matrix physics to control cell fate has been a promising way for cell and tissue engineering. However, the nanoscale situations taking place during the topography-regulated cell-matrix interactions are still not fully understood to the best of our knowledge. The invention of atomic force microscopy (AFM) provides a powerful tool to characterize the structures and properties of living biological systems under aqueous conditions with unprecedented spatial resolution. In this work, with the use of AFM, structural and mechanical dynamics of individual cells grown on micro-/nanotopographical surface were revealed. First, the microgroove patterned silicon substrates were fabricated by photolithography. Next, nanogranular topography was formed on microgroove substrates by cell culture medium protein deposition, which was visualized by in situ AFM imaging. The micro-/nanotopographical substrates were then used to grow two types of cells (3T3 cell or MCF-7 cell). AFM morphological imaging and mechanical measurements were applied to characterize the changes of cells grown on the micro-/nanotopographical substrates. The experimental results showed the significant alterations in cellular structures and cellular mechanics caused by micro-/nanotopography. The study provides a novel way based on AFM to unveil the native nanostructures and mechanical properties of cell-matrix interfaces with high spatial resolution in liquids, which will have potential impacts on the studies of topography-tuned cell behaviors.
Collapse
|
19
|
Micalet A, Moeendarbary E, Cheema U. 3D In Vitro Models for Investigating the Role of Stiffness in Cancer Invasion. ACS Biomater Sci Eng 2021. [PMID: 34081437 DOI: 10.1021/acsbiomaterials.0c01530] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Tumorigenesis is attributed to the interactions of cancer cells with the tumor microenvironment through both biochemical cues and physical stimuli. Increased matrix deposition and realignment of the collagen fibers are detected by cancer cells, inducing epithelial-to-mesenchymal transition, which in turn stimulates cell motility and invasiveness. METHODS This review provides an overview of current research on the role of the physical microenvironment in cancer invasion. This was achieved by using a systematic approach and providing meta-analyses. Particular focus was placed on in vitro three-dimensional models of epithelial cancers. We investigated questions such as the effect of matrix stiffening, activation of stromal cells, and identified potential advances in mechano-based therapies. RESULTS Meta-analysis revealed that 64% of studies report cancer invasion promotion as stiffness increases, while 36% report the opposite. Experimental approaches and data interpretations were varied, each affecting the invasion of cancer differently. Examples are the experimental timeframes used (24 h to 21 days), the type of polymer used (24 types), and choice of cell line (33 cell lines). The stiffness of the 3D matrices varied from 0.5 to 300 kPa and 19% of these matrices' stiffness were outside commonly accepted physiological range. 100% of the studies outside biological stiffness range (above 20 kPa) report that stiffness does not promote cancer invasion. CONCLUSIONS Taking this analysis into account, we inform on the type of experimental approaches that could be the most relevant and provide what would be a standardized protocol and reporting strategy.
Collapse
Affiliation(s)
- Auxtine Micalet
- Department of Mechanical Engineering, University College London (UCL), Torrington Place, London, U.K. WC1E 6BT.,Division of Surgery and Interventional Sciences, UCL Centre for 3D Models of Health and Disease, University College London (UCL), Charles Bell House, London, U.K. W1W 7TS
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London (UCL), Torrington Place, London, U.K. WC1E 6BT.,Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, Massachusetts 02139, United States
| | - Umber Cheema
- Division of Surgery and Interventional Sciences, UCL Centre for 3D Models of Health and Disease, University College London (UCL), Charles Bell House, London, U.K. W1W 7TS
| |
Collapse
|
20
|
Rheinlaender J, Wirbel H, Schäffer TE. Spatial correlation of cell stiffness and traction forces in cancer cells measured with combined SICM and TFM. RSC Adv 2021; 11:13951-13956. [PMID: 35423943 PMCID: PMC8697701 DOI: 10.1039/d1ra01277k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/06/2021] [Indexed: 12/31/2022] Open
Abstract
The mechanical properties of cancer cells at the single-cell and the subcellular level might be the key for answering long-standing questions in the diagnosis and treatment of cancer. However, the subcellular distribution of two main mechanical properties, cell stiffness and traction forces, has been investigated only rarely and qualitatively yet. Here, we present the first direct combination of scanning ion conductance microscopy (SICM) and traction force microscopy (TFM), which we used to identify a correlation between the local stiffness and the local traction force density in living cells. We found a correlation in normal breast epithelial cells, but no correlation in cancerous breast epithelial cells. This indicates that the interplay between cell stiffness and traction forces is altered in cancer cells as compared to healthy cells, which might give new insight in the research field of cancer cell mechanobiology.
Collapse
Affiliation(s)
- Johannes Rheinlaender
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| | - Hannes Wirbel
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| | - Tilman E Schäffer
- Institute of Applied Physics, University of Tübingen Auf der Morgenstelle 10 72076 Tübingen Germany +49 7071 29 5093 +49 7071 29 76030
| |
Collapse
|
21
|
Cell Cytoskeleton and Stiffness Are Mechanical Indicators of Organotropism in Breast Cancer. BIOLOGY 2021; 10:biology10040259. [PMID: 33805866 PMCID: PMC8064360 DOI: 10.3390/biology10040259] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/05/2021] [Accepted: 03/06/2021] [Indexed: 12/23/2022]
Abstract
Simple Summary Cancer cell dissemination exhibits organ preference or organotropism. Although the influence of intrinsic biochemical factors on organotropism has been intensely studied, little is known about the roles of mechanical properties of metastatic cancer cells. Our study suggests that there may be a correlation between cell cytoskeleton/stiffness and organotropism. We find that the cytoskeleton and stiffness of breast cancer cell subpopulations with different metastatic preference match the mechanics of the metastasized organs. The modification of cell cytoskeleton significantly influences the organotropism-related gene expression pattern and mechanoresponses on soft substrates which mimic brain tissue stiffness. These findings highlight the key role of cell cytoskeleton in specific organ metastasis, which may not only reflect but also impact the metastatic organ preference. Abstract Tumor metastasis involves the dissemination of tumor cells from the primary lesion to other organs and the subsequent formation of secondary tumors, which leads to the majority of cancer-related deaths. Clinical findings show that cancer cell dissemination is not random but exhibits organ preference or organotropism. While intrinsic biochemical factors of cancer cells have been extensively studied in organotropism, much less is known about the role of cell cytoskeleton and mechanics. Herein, we demonstrate that cell cytoskeleton and mechanics are correlated with organotropism. The result of cell stiffness measurements shows that breast cancer cells with bone tropism are much stiffer with enhanced F-actin, while tumor cells with brain tropism are softer with lower F-actin than their parental cells. The difference in cellular stiffness matches the difference in the rigidity of their metastasized organs. Further, disrupting the cytoskeleton of breast cancer cells with bone tropism not only elevates the expressions of brain metastasis-related genes but also increases cell spreading and proliferation on soft substrates mimicking the stiffness of brain tissue. Stabilizing the cytoskeleton of cancer cells with brain tropism upregulates bone metastasis-related genes while reduces the mechanoadaptation ability on soft substrates. Taken together, these findings demonstrate that cell cytoskeleton and biophysical properties of breast cancer subpopulations correlate with their metastatic preference in terms of gene expression pattern and mechanoadaptation ability, implying the potential role of cell cytoskeleton in organotropism.
Collapse
|
22
|
Amirghasemi F, Adjei-Sowah E, Pockaj BA, Nikkhah M. Microengineered 3D Tumor Models for Anti-Cancer Drug Discovery in Female-Related Cancers. Ann Biomed Eng 2021; 49:1943-1972. [PMID: 33403451 DOI: 10.1007/s10439-020-02704-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
The burden of cancer continues to increase in society and negatively impacts the lives of numerous patients. Due to the high cost of current treatment strategies, there is a crucial unmet need to develop inexpensive preclinical platforms to accelerate the process of anti-cancer drug discovery to improve outcomes in cancer patients, most especially in female patients. Many current methods employ expensive animal models which not only present ethical concerns but also do not often accurately predict human physiology and the outcomes of anti-cancer drug responsiveness. Conventional treatment approaches for cancer generally include systemic therapy after a surgical procedure. Although this treatment technique is effective, the outcome is not always positive due to various complex factors such as intratumor heterogeneity and confounding factors within the tumor microenvironment (TME). Patients who develop metastatic disease still have poor prognosis. To that end, recent efforts have attempted to use 3D microengineered platforms to enhance the predictive power and efficacy of anti-cancer drug screening, ultimately to develop personalized therapies. Fascinating features of microengineered assays, such as microfluidics, have led to the advancement in the development of the tumor-on-chip technology platforms, which have shown tremendous potential for meaningful and physiologically relevant anti-cancer drug discovery and screening. Three dimensional microscale models provide unprecedented ability to unveil the biological complexities of cancer and shed light into the mechanism of anti-cancer drug resistance in a timely and resource efficient manner. In this review, we discuss recent advances in the development of microengineered tumor models for anti-cancer drug discovery and screening in female-related cancers. We specifically focus on female-related cancers to draw attention to the various approaches being taken to improve the survival rate of women diagnosed with cancers caused by sex disparities. We also briefly discuss other cancer types like colon adenocarcinomas and glioblastoma due to their high rate of occurrence in females, as well as the high likelihood of sex-biased mutations which complicate current treatment strategies for women. We highlight recent advances in the development of 3D microscale platforms including 3D tumor spheroids, microfluidic platforms as well as bioprinted models, and discuss how they have been utilized to address major challenges in the process of drug discovery, such as chemoresistance, intratumor heterogeneity, drug toxicity, etc. We also present the potential of these platform technologies for use in high-throughput drug screening approaches as a replacements of conventional assays. Within each section, we will provide our perspectives on advantages of the discussed platform technologies.
Collapse
Affiliation(s)
- Farbod Amirghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA
| | - Emmanuela Adjei-Sowah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA
| | - Barbara A Pockaj
- Division of Surgical Oncology and Endocrine Surgery, Department of Surgery, Mayo Clinic, Phoenix, AZ, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287-9709, USA. .,Biodesign Virginia G. Piper Center for Personalized Diagnostics, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
23
|
Azadi S, Tafazzoli Shadpour M, Warkiani ME. Characterizing the effect of substrate stiffness on the extravasation potential of breast cancer cells using a 3D microfluidic model. Biotechnol Bioeng 2020; 118:823-835. [PMID: 33111314 DOI: 10.1002/bit.27612] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/02/2020] [Accepted: 10/20/2020] [Indexed: 12/14/2022]
Abstract
Different biochemical and biomechanical cues from tumor microenvironment affect the extravasation of cancer cells to distant organs; among them, the mechanical signals are poorly understood. Although the effect of substrate stiffness on the primary migration of cancer cells has been previously probed, its role in regulating the extravasation ability of cancer cells is still vague. Herein, we used a microfluidic device to mimic the extravasation of tumor cells in a 3D microenvironment containing cancer cells, endothelial cells, and the biological matrix. The microfluidic-based extravasation model was utilized to probe the effect of substrate stiffness on the invasion ability of breast cancer cells. MCF7 and MDA-MB-231 cancer cells were cultured among substrates with different stiffness which followed by monitoring their extravasation capability through the microfluidic device. Our results demonstrated that acidic collagen at a concentration of 2.5 mg/ml promotes migration of cancer cells. Additionally, the substrate softening resulted in up to 46% reduction in the invasion of breast cancer cells. The substrate softening not only affected the number of extravasated cells but also reduced their migration distance up to 53%. We further investigated the secreted level of matrix metalloproteinase 9 (MMP9) and identified that there is a positive correlation between substrate stiffening, MMP9 concentration, and extravasation of cancer cells. These findings suggest that the substrate stiffness mediates the cancer cells extravasation in a microfluidic model. Changes in MMP9 level could be one of the possible underlying mechanisms which need more investigations to be addressed thoroughly.
Collapse
Affiliation(s)
- Shohreh Azadi
- Faculty of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | | | - Majid E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia.,Institute of Molecular Medicine, Sechenov University, Moscow, Russia
| |
Collapse
|
24
|
Suter N, Stebel S, Rianna C, Radmacher M, Brüggemann D. Spatial patterning of nanofibrous collagen scaffolds modulates fibroblast morphology. Biofabrication 2020; 13:015007. [DOI: 10.1088/1758-5090/abb744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
25
|
Northcutt LA, Suarez-Arnedo A, Rafat M. Emerging Biomimetic Materials for Studying Tumor and Immune Cell Behavior. Ann Biomed Eng 2020; 48:2064-2077. [PMID: 31617045 PMCID: PMC7156320 DOI: 10.1007/s10439-019-02384-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 10/09/2019] [Indexed: 02/06/2023]
Abstract
Cancer is one of the leading causes of death both in the United States and worldwide. The dynamic microenvironment in which tumors grow consists of fibroblasts, immune cells, extracellular matrix (ECM), and cytokines that enable progression and metastasis. Novel biomaterials that mimic these complex surroundings give insight into the biological, chemical, and physical environment that cause cancer cells to metastasize and invade into other tissues. Two-dimensional (2D) cultures are useful for gaining limited information about cancer cell behavior; however, they do not accurately represent the environments that cells experience in vivo. Recent advances in the design and tunability of diverse three-dimensional (3D) biomaterials complement biological knowledge and allow for improved recapitulation of in vivo conditions. Understanding cell-ECM and cell-cell interactions that facilitate tumor survival will accelerate the design of more effective therapies. This review discusses innovative materials currently being used to study tumor and immune cell behavior and interactions, including materials that mimic the ECM composition, mechanical stiffness, and integrin binding sites of the tumor microenvironment.
Collapse
Affiliation(s)
- Logan A Northcutt
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
| | | | - Marjan Rafat
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA.
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Engineering and Science Building, Rm. 426, Nashville, TN, 37212, USA.
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
26
|
Zanotelli MR, Chada NC, Johnson CA, Reinhart-King CA. The Physical Microenvironment of Tumors: Characterization and Clinical Impact. ACTA ACUST UNITED AC 2020. [DOI: 10.1142/s1793048020300029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The tumor microenvironment plays a critical role in tumorigenesis and metastasis. As tightly controlled extracellular matrix homeostasis is lost during tumor progression, a dysregulated extracellular matrix can significantly alter cellular phenotype and drive malignancy. Altered physical properties of the tumor microenvironment alter cancer cell behavior, limit delivery and efficacy of therapies, and correlate with tumorigenesis and patient prognosis. The physical features of the extracellular matrix during tumor progression have been characterized; however, a wide range of methods have been used between studies and cancer types resulting in a large range of reported values. Here, we discuss the significant mechanical and structural properties of the tumor microenvironment, summarizing their reported values and clinical impact across cancer type and grade. We attempt to integrate the values in the literature to identify sources of reported differences and commonalities to better understand how aberrant extracellular matrix dynamics contribute to cancer progression. An intimate understanding of altered matrix properties during malignant transformation will be crucial in effectively detecting, monitoring, and treating cancer.
Collapse
Affiliation(s)
- Matthew R. Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Weill Hall, Ithaca, NY 14583, USA
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Avenue, Nashville, TN 37235, USA
| | - Neil C. Chada
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Avenue, Nashville, TN 37235, USA
| | - C. Andrew Johnson
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Avenue, Nashville, TN 37235, USA
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Avenue, Nashville, TN 37235, USA
| |
Collapse
|
27
|
Criado-Gonzalez M, Loftin B, Rodon Fores J, Vautier D, Kocgozlu L, Jierry L, Schaaf P, Boulmedais F, Harth E. Enzyme assisted peptide self-assemblies trigger cell adhesion in high density oxime based host gels. J Mater Chem B 2020; 8:4419-4427. [PMID: 32186320 DOI: 10.1039/d0tb00456a] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Peptide supramolecular self-assemblies are recognized as important components in responsive hydrogel based materials with applications in tissue engineering and regenerative medicine. Studying the influence of hydrogel matrices on the self-assembly behavior of peptides and interaction with cells is essential to guide the future development of engineered biomaterials. In this contribution, we present a PEG based host hydrogel material generated by oxime click chemistry that shows cellular adhesion behavior in response to enzyme assisted peptide self-assembly (EASA) within the host gel. This hydrogel prepared from poly(dimethylacrylamide-co-diacetoneacrylamide), poly(DMA-DAAM) with high molar fractions (49%) of DAAM and dialkoxyamine PEG cross-linker, was studied in the presence of embedded enzyme alkaline phosphatase (AP) and a non-adhesive cell behavior towards NIH 3T3 fibroblasts was observed. When brought into contact with a Fmoc-FFpY peptide solution (pY: phosphorylated tyrosine), the gel forms intercalated Fmoc-FFY peptide self-assemblies upon diffusion of Fmoc-FFpY into the cross-linked hydrogel network as was confirmed by circular dichroism, fluorescence emission spectroscopy and confocal microscopy. Nevertheless, the mechanical properties do not change significantly after the peptide self-assembly in the host gel. This enzyme assisted peptide self-assembly promotes fibroblast cell adhesion that can be enhanced if Fmoc-F-RGD peptides are added to the pre-gelator Fmoc-FFpY peptide solution. Cell adhesion results mainly from interactions of cells with the non-covalent peptide self-assemblies present in the gel despite the fact that the mechanical properties are very close to those of the native host gel. This result is in contrast to numerous studies which showed that the mechanical properties of a substrate are key parameters of cell adhesion. It opens up the possibility to develop a diverse set of hybrid materials to control cell fate in culture due to tailored self-assemblies of peptides responding to the environment provided by the host guest gel.
Collapse
Affiliation(s)
- Miryam Criado-Gonzalez
- Université de Strasbourg, CNRS, Institut Charles Sadron UPR 22, 67034 Strasbourg, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
de Sousa JS, Freire RS, Sousa FD, Radmacher M, Silva AFB, Ramos MV, Monteiro-Moreira ACO, Mesquita FP, Moraes MEA, Montenegro RC, Oliveira CLN. Double power-law viscoelastic relaxation of living cells encodes motility trends. Sci Rep 2020; 10:4749. [PMID: 32179816 PMCID: PMC7075927 DOI: 10.1038/s41598-020-61631-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
Living cells are constantly exchanging momentum with their surroundings. So far, there is no consensus regarding how cells respond to such external stimuli, although it reveals much about their internal structures, motility as well as the emergence of disorders. Here, we report that twelve cell lines, ranging from healthy fibroblasts to cancer cells, hold a ubiquitous double power-law viscoelastic relaxation compatible with the fractional Kelvin-Voigt viscoelastic model. Atomic Force Microscopy measurements in time domain were employed to determine the mechanical parameters, namely, the fast and slow relaxation exponents, the crossover timescale between power law regimes, and the cell stiffness. These cell-dependent quantities show strong correlation with their collective migration and invasiveness properties. Beyond that, the crossover timescale sets the fastest timescale for cells to perform their biological functions.
Collapse
Affiliation(s)
- J S de Sousa
- Departamento de Física, Universidade Federal do Ceará, 60455-970, Fortaleza, Ceará, Brazil.
| | - R S Freire
- Central Analítica, Universidade Federal do Ceará, 60455-970, Fortaleza, Ceará, Brazil
| | - F D Sousa
- Departamento de Física, Universidade Federal do Ceará, 60455-970, Fortaleza, Ceará, Brazil
| | - M Radmacher
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, 28359, Bremen, Germany
| | - A F B Silva
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, 60440-554, Fortaleza, Ceará, Brazil
| | - M V Ramos
- Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Ceará, 60440-554, Fortaleza, Ceará, Brazil
| | - A C O Monteiro-Moreira
- Centro de Biologia Experimental, Universidade de Fortaleza, 60811-905, Fortaleza, Ceará, Brazil
| | - F P Mesquita
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Universidade Federal do Ceará, 60430-275, Fortaleza, Ceará, Brazil
| | - M E A Moraes
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Universidade Federal do Ceará, 60430-275, Fortaleza, Ceará, Brazil
| | - R C Montenegro
- Núcleo de Pesquisa e Desenvolvimento de Medicamentos, Universidade Federal do Ceará, 60430-275, Fortaleza, Ceará, Brazil
| | - C L N Oliveira
- Departamento de Física, Universidade Federal do Ceará, 60455-970, Fortaleza, Ceará, Brazil
| |
Collapse
|
29
|
Xu Y, Xiao L, Chang Y, Cao Y, Chen C, Wang D. pH and Redox Dual-Responsive MSN-S-S-CS as a Drug Delivery System in Cancer Therapy. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E1279. [PMID: 32178282 PMCID: PMC7143049 DOI: 10.3390/ma13061279] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/09/2020] [Accepted: 03/09/2020] [Indexed: 01/23/2023]
Abstract
In order to achieve a controlled release drug delivery system (DDS) for cancer therapy, a pH and redox dual-responsive mesoporous silica nanoparticles (MSN)-sulfur (S)-S- chitosan (CS) DDS was prepared via an amide reaction of dithiodipropionic acid with amino groups on the surface of MSN and amino groups on the surface of CS. Using salicylic acid (SA) as a model drug, SA@MSN-S-S-CS was prepared by an impregnation method. Subsequently, the stability, swelling properties and drug release properties of the DDS were studied by x-ray diffraction, scanning electron microscopy, Fourier transform infrared microspectroscopy, size and zeta potential as well as Brunauer-Emmett-Teller surface area. Pore size and volume of the composites decreased after drug loading but maintained a stable structure. The calculated drug loading rate and encapsulation efficiency were 8.17% and 55.64%, respectively. The in vitro drug release rate was 21.54% in response to glutathione, and the release rate showed a marked increase as the pH decreased. Overall, double response functions of MSN-S-S-CS had unique advantages in controlled drug delivery, and may be a new clinical application of DDS in cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Yuan Cao
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China; (Y.X.); (L.X.); (Y.C.)
| | - Changguo Chen
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China; (Y.X.); (L.X.); (Y.C.)
| | - Dan Wang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 401331, China; (Y.X.); (L.X.); (Y.C.)
| |
Collapse
|
30
|
Mirastschijski U, Schwab I, Coger V, Zier U, Rianna C, He W, Maedler K, Kelm S, Radtke A, Belge G, Lindner P, Stahl F, Scharpenberg M, Lasota L, Timm J. Lung Surfactant Accelerates Skin Wound Healing: A Translational Study with a Randomized Clinical Phase I Study. Sci Rep 2020; 10:2581. [PMID: 32054903 PMCID: PMC7018835 DOI: 10.1038/s41598-020-59394-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/28/2020] [Indexed: 01/10/2023] Open
Abstract
Lung surfactants are used for reducing alveolar surface tension in preterm infants to ease breathing. Phospholipid films with surfactant proteins regulate the activity of alveolar macrophages and reduce inflammation. Aberrant skin wound healing is characterized by persistent inflammation. The aim of the study was to investigate if lung surfactant can promote wound healing. Preclinical wound models, e.g. cell scratch assays and full-thickness excisional wounds in mice, and a randomized, phase I clinical trial in healthy human volunteers using a suction blister model were used to study the effect of the commercially available bovine lung surfactant on skin wound repair. Lung surfactant increased migration of keratinocytes in a concentration-dependent manner with no effect on fibroblasts. Significantly reduced expression levels were found for pro-inflammatory and pro-fibrotic genes in murine wounds. Because of these beneficial effects in preclinical experiments, a clinical phase I study was initiated to monitor safety and tolerability of surfactant when applied topically onto human wounds and normal skin. No adverse effects were observed. Subepidermal wounds healed significantly faster with surfactant compared to control. Our study provides lung surfactant as a strong candidate for innovative treatment of chronic skin wounds and as additive for treatment of burn wounds to reduce inflammation and prevent excessive scarring.
Collapse
Affiliation(s)
- Ursula Mirastschijski
- Center for Biomolecular Interactions Bremen, Faculty of Biology and Chemistry, University of Bremen, Bremen, Germany.
| | - Igor Schwab
- Department of Plastic, Reconstructive and Aesthetic Surgery, Klinikum Bremen-Mitte, Bremen, Germany
| | - Vincent Coger
- Department of Experimental Plastic Surgery, Kerstin Reimers Laboratory for Regeneration Biology, Hannover Medical School, Hannover, Germany
| | - Ulrich Zier
- Center for Biomolecular Interactions Bremen, Faculty of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Carmela Rianna
- Institute of Biophysics, University of Bremen, Bremen, Germany
| | - Wei He
- Center for Biomolecular Interactions Bremen, Faculty of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Kathrin Maedler
- Center for Biomolecular Interactions Bremen, Faculty of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Sørge Kelm
- Center for Biomolecular Interactions Bremen, Faculty of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Arlo Radtke
- Faculty of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Gazanfer Belge
- Faculty of Biology and Chemistry, University of Bremen, Bremen, Germany
| | - Patrick Lindner
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Frank Stahl
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | - Martin Scharpenberg
- University of Bremen, Competence Center for Clinical Trials Bremen, Bremen, Germany
| | - Lukas Lasota
- University of Bremen, Competence Center for Clinical Trials Bremen, Bremen, Germany
| | - Jürgen Timm
- University of Bremen, Competence Center for Clinical Trials Bremen, Bremen, Germany
| |
Collapse
|
31
|
Rianna C, Radmacher M, Kumar S. Direct evidence that tumor cells soften when navigating confined spaces. Mol Biol Cell 2020; 31:1726-1734. [PMID: 31995446 PMCID: PMC7521845 DOI: 10.1091/mbc.e19-10-0588] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The mechanical properties of cells strongly regulate many physiological and pathological processes. For example, in cancer, invasive and metastatic tumor cells have often been reported to be softer than nontumor cells, raising speculation that cancer cells might adaptively soften to facilitate migration through narrow tissue spaces. Despite growing interest in targeting cell softening to impede invasion and metastasis, it remains to be directly demonstrated that tumor cells soften as they migrate through confined spaces. Here, we address this open question by combining topographically patterned substrates with atomic force microscopy (AFM). Using a polydimethylsiloxane open-roof microdevice featuring tapered, fibronectin-coated channels, we followed the migration of U2OS cells through various stages of confinement while simultaneously performing AFM indentation. As cells progress from unconfined migration to fully confined migration, cells soften and exclude Yes-associated protein from the nucleus. Superresolution imaging reveals that confinement induces remodeling of actomyosin stress fiber architecture. Companion studies with flat one-dimensional microlines indicate that the changes in cytoarchitecture and mechanics are intrinsically driven by topographical confinement rather than changes in cellular aspect ratio. Our studies represent among the most direct evidence to date that tumor cells soften during confined migration and support cell softening as a mechanoadaptive mechanism during invasion.
Collapse
Affiliation(s)
- Carmela Rianna
- Institute of Biophysics, University of Bremen, 28359 Bremen, Germany.,Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720
| | - Manfred Radmacher
- Institute of Biophysics, University of Bremen, 28359 Bremen, Germany
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California 94720
| |
Collapse
|
32
|
Chen M, Zeng J, Ruan W, Zhang Z, Wang Y, Xie S, Wang Z, Yang H. Examination of the relationship between viscoelastic properties and the invasion of ovarian cancer cells by atomic force microscopy. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2020; 11:568-582. [PMID: 32318318 PMCID: PMC7155897 DOI: 10.3762/bjnano.11.45] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 03/04/2020] [Indexed: 05/17/2023]
Abstract
The mechanical properties of cells could serve as an indicator for disease progression and early cancer diagnosis. This study utilized atomic force microscopy (AFM) to measure the viscoelastic properties of ovarian cancer cells and then examined the association with the invasion of ovarian cancer at the level of living single cells. Elasticity and viscosity of the ovarian cancer cells OVCAR-3 and HO-8910 are significantly lower than those of the human ovarian surface epithelial cell (HOSEpiC) control. Further examination found a dramatic increase of migration/invasion and an obvious decease of microfilament density in OVCAR-3 and HO-8910 cells. Also, there was a significant relationship between viscoelastic and biological properties among these cells. In addition, the elasticity was significantly increased in OVCAR-3 and HO-8910 cells after the treatment with the anticancer compound echinomycin (Ech), while no obvious change was found in HOSEpiC cells after Ech treatment. Interestingly, Ech seemed to have no effect on the viscosity of the cells. Ech significantly inhibited the migration/invasion and significantly increased the microfilament density in OVCAR-3 and HO-8910 cells, which was significantly related with the elasticity of the cells. An increase of elasticity and a decrease of invasion were found in OVCAR-3 and HO-8910 cells after Ech treatment. Together, this study clearly demonstrated the association of viscoelastic properties with the invasion of ovarian cancer cells and shed a light on the biomechanical changes for early diagnosis of tumor transformation and progression at single-cell level.
Collapse
Affiliation(s)
- Mengdan Chen
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Jinshu Zeng
- Department of Ultrasound Medical, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Weiwei Ruan
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Zhenghong Zhang
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Yuhua Wang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Shusen Xie
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| | - Zhengchao Wang
- Fujian Provincial Key Laboratory for Developmental Biology and Neurosciences, College of Life Sciences, Fujian Normal University, Fuzhou 350007, China
| | - Hongqin Yang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory for Photonics Technology, Fujian Normal University, Fuzhou 350007, China
| |
Collapse
|
33
|
Lam BP, Cheung SKC, Lam YW, Pang SW. Microenvironmental topographic cues influence migration dynamics of nasopharyngeal carcinoma cells from tumour spheroids. RSC Adv 2020; 10:28975-28983. [PMID: 35520045 PMCID: PMC9055862 DOI: 10.1039/d0ra03740k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
Tumour metastasis is a complex process that strongly influences the prognosis and treatment of cancer. Apart from intracellular factors, recent studies have indicated that metastasis also depends on microenvironmental factors such as the biochemical, mechanical and topographical properties of the surrounding extracellular matrix (ECM) of tumours. In this study, as a proof of concept, we conducted tumour spheroid dissemination assay on engineered surfaces with micrograting patterns. Nasopharyngeal spheroids were generated by the 3D culture of nasopharyngeal carcinoma (NPC43) cells, a newly established cell line that maintains a high level of Epstein–Barr virus, a hallmark of NPC. Three types of collagen I-coated polydimethylsiloxane (PDMS) substrates were used, with 15 μm deep “trenches” that grated the surfaces: (a) 40/10 μm ridges (R)/trenches (T), (b) 18/18 μm (R/T) and (c) 50/50 μm (R/T). The dimensions of these patterns were designed to test how various topographical cues, different with respect to the size of tumour spheroids and individual NPC43 cells, might affect dissemination behaviours. Spreading efficiencies on all three patterned surfaces, especially 18/18 μm (R/T), were lower than that on flat PDMS surface. The outspreading cell sheets on flat and 40/10 μm (R/T) surfaces were relatively symmetrical but appeared ellipsoid and aligned with the main axes of the 18/18 μm (R/T) and 50/50 μm (R/T) grating platforms. Focal adhesions (FAs) were found to preferentially formed on the ridges of all patterns. The number of FAs per spheroid was strongly influenced by the grating pattern, with the least FAs on the 40/10 μm (R/T) and the most on the 50/50 μm (R/T) substrate. Taken together, these data indicate a previously unknown effect of surface topography on the efficiency and directionality of cancer cell spreading from tumour spheroids, suggesting that topography, like ECM biochemistry and stiffness, can influence the migration dynamics in 3D cell culture models. Investigation of collective migration of nasopharyngeal carcinoma cells from tumour spheroids on micro-engineered platforms that induced asymmetrical tumour shape.![]()
Collapse
Affiliation(s)
- Bowie P. Lam
- Department of Electrical Engineering
- City University of Hong Kong
- Hong Kong
- Centre for Biosystems, Neuroscience, and Nanotechnology
- City University of Hong Kong
| | - Sarah K. C. Cheung
- Centre for Biosystems, Neuroscience, and Nanotechnology
- City University of Hong Kong
- Hong Kong
- Department of Chemistry
- City University of Hong Kong
| | - Yun W. Lam
- Centre for Biosystems, Neuroscience, and Nanotechnology
- City University of Hong Kong
- Hong Kong
- Department of Chemistry
- City University of Hong Kong
| | - Stella W. Pang
- Department of Electrical Engineering
- City University of Hong Kong
- Hong Kong
- Centre for Biosystems, Neuroscience, and Nanotechnology
- City University of Hong Kong
| |
Collapse
|
34
|
Alvarez-Paino M, Amer MH, Nasir A, Cuzzucoli Crucitti V, Thorpe J, Burroughs L, Needham D, Denning C, Alexander MR, Alexander C, Rose FRAJ. Polymer Microparticles with Defined Surface Chemistry and Topography Mediate the Formation of Stem Cell Aggregates and Cardiomyocyte Function. ACS APPLIED MATERIALS & INTERFACES 2019; 11:34560-34574. [PMID: 31502820 DOI: 10.1021/acsami.9b04769] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Surface-functionalized microparticles are relevant to fields spanning engineering and biomedicine, with uses ranging from cell culture to advanced cell delivery. Varying topographies of biomaterial surfaces are also being investigated as mediators of cell-material interactions and subsequent cell fate. To investigate competing or synergistic effects of chemistry and topography in three-dimensional cell cultures, methods are required to introduce these onto microparticles without modification of their underlying morphology or bulk properties. In this study, a new approach for surface functionalization of poly(lactic acid) (PLA) microparticles is reported that allows decoration of the outer shell of the polyesters with additional polymers via aqueous atom transfer radical polymerization routes. PLA microparticles with smooth or dimpled surfaces were functionalized with poly(poly(ethylene glycol) methacrylate) and poly[N-(3-aminopropyl)methacrylamide] brushes, chosen for their potential abilities to mediate cell adhesion. X-ray photoelectron spectroscopy and time-of-flight secondary ion mass spectrometry analysis indicated homogeneous coverage of the microparticles with polymer brushes while maintaining the original topographies. These materials were used to investigate the relative importance of surface chemistry and topography both on the formation of human immortalized mesenchymal stem cell (hiMSCs) particle-cell aggregates and on the enhanced contractility of cardiomyocytes derived from human-induced pluripotent stem cells (hiPSC-CMs). The influence of surface chemistry was found to be more important on the size of particle-cell aggregates than topographies. In addition, surface chemistries that best promoted hiMSC attachment also improved hiPSC-CM attachment and contractility. These studies demonstrated a new route to obtain topo-chemical combinations on polyester-based biomaterials and provided clear evidence for the predominant effect of surface functionality over micron-scale dimpled topography in cell-microparticle interactions. These findings, thus, provide new guiding principles for the design of biomaterial interfaces to direct cell function.
Collapse
|
35
|
Viji Babu PK, Rianna C, Mirastschijski U, Radmacher M. Nano-mechanical mapping of interdependent cell and ECM mechanics by AFM force spectroscopy. Sci Rep 2019; 9:12317. [PMID: 31444369 PMCID: PMC6707266 DOI: 10.1038/s41598-019-48566-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
Extracellular matrix (ECM), as a dynamic component of the tissue, influences cell behavior and plays an important role in cell mechanics and tissue homeostasis. Reciprocally, this three-dimensional scaffold is dynamically, structurally and mechanically modified by cells. In the field of biophysics, the independent role of cell and ECM mechanics has been largely investigated; however, there is a lack of experimental data reporting the interdependent interplay between cell and ECM mechanics, measured simultaneously. Here, using Atomic Force Microscopy (AFM) we have characterized five different decellularized matrices diverse in their topography, ECM composition and stiffness and cultured them with normal and pathological fibroblasts (scar and Dupuytren's). We investigated the change in topography and elasticity of these matrices due to cell seeding, by using AFM peak force imaging and mechanical mapping, respectively. We found normal fibroblasts soften these matrices more than pathological fibroblasts, suggesting that pathological fibroblasts are profoundly influencing tissue stiffening in fibrosis. We detected different ECM composition of decellularized matrices used here influences fibroblast stiffness, thus highlighting that cell mechanics not only depends on ECM stiffness but also on their composition. We used confocal microscopy to assess fibroblasts invasion and found pathological fibroblasts were invading the matrices deeper than normal fibroblasts.
Collapse
Affiliation(s)
| | - Carmela Rianna
- Institute of Biophysics, University of Bremen, Bremen, Germany
| | - Ursula Mirastschijski
- Wound Repair Unit, Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | | |
Collapse
|
36
|
Cardiac Fibroblast to Myofibroblast Phenotype Conversion-An Unexploited Therapeutic Target. J Cardiovasc Dev Dis 2019; 6:jcdd6030028. [PMID: 31426390 PMCID: PMC6787657 DOI: 10.3390/jcdd6030028] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 02/07/2023] Open
Abstract
Fibrosis occurs when the synthesis of extracellular matrix outpaces its degradation, and over time can negatively impact tissue and organ function. In the case of cardiac fibrosis, contraction and relaxation of the heart can be impaired to the point of precipitating heart failure, while at the same time fibrosis can result in arrhythmias due to altered electrical properties of the myocardium. The critical event in the evolution of cardiac fibrosis is the phenotype conversion of cardiac fibroblasts to their overly-active counterparts, myofibroblasts: cells demarked by their expression of novel markers such as periostin, by their gain of contractile activity, and by their pronounced and prolonged increase in the production of extracellular matrix components such as collagens. The phenotype change is dramatic, and can be triggered by many stimuli, including mechanical force, inflammatory cytokines, and growth factors. This review will explore fibroblast to myofibroblast transition mechanisms and will consider the therapeutic potential of targeting this process as a means to arrest or even reverse cardiac fibrosis.
Collapse
|
37
|
Azadi S, Aboulkheyr Es H, Razavi Bazaz S, Thiery JP, Asadnia M, Ebrahimi Warkiani M. Upregulation of PD-L1 expression in breast cancer cells through the formation of 3D multicellular cancer aggregates under different chemical and mechanical conditions. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:118526. [PMID: 31398408 DOI: 10.1016/j.bbamcr.2019.118526] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/20/2019] [Accepted: 08/04/2019] [Indexed: 12/30/2022]
Abstract
Expression of programmed death-ligand 1 (PD-L1) in cancer cells plays an important role in cancer-immune cell interaction. The emerging evidence suggests regulation of PD-L1 expression by several tumor microenvironmental cues. However, the association of PD-L1 expression with chemical and mechanical features of the tumor microenvironment, specifically epidermal growth factor receptor (EGFR) signaling and matrix stiffness, remains elusive. Herein, we determine whether EGFR targeting and substrate stiffness affect the regulation of PD-L1 expression. Breast carcinoma cell lines, MCF7 and MDA-MB-231, were cultured under different conditions targeting EGFR and exposing cells to distinct substrate stiffness to evaluate PD-L1 expression. Furthermore, the ability to form aggregates in short-term culture of breast carcinoma cells and its effect on expression level of PD-L1 was probed. Our results indicated that PD-L1 expression was altered in response to both EGFR inhibition and substrate stiffness. Additionally, a positive association between the formation of multicellular aggregates and PD-L1 expression was observed. MDA-MB-231 cells expressed the highest PD-L1 level on a stiff substrate, while inhibition of EGFR reduced expression of PD-L1. The results suggested that both physical and chemical features of tumor microenvironment regulate PD-L1 expression through alteration of tumor aggregate formation potential. In line with these results, the in-silico study highlighted a positive correlation between PD-L1 expression, EGFR signaling, epithelial to mesenchymal transition related transcription factors (EMT-TFs) and stemness markers in metastatic breast cancer. These findings improve our understanding of regulation of PD-L1 expression by tumor microenvironment leading to evasion of tumor cells from the immune system.
Collapse
Affiliation(s)
- Shohreh Azadi
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia; School of Engineering, Macquarie University, Sydney 2109, Australia
| | - Hamidreza Aboulkheyr Es
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Sajad Razavi Bazaz
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Jean Paul Thiery
- Inserm Unit 1186, Comprehensive Cancer Center, Institut Gustave Roussy, Villejuif, France
| | - Mohsen Asadnia
- School of Engineering, Macquarie University, Sydney 2109, Australia
| | - Majid Ebrahimi Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia; Institute of Molecular Medicine, Sechenov University, Moscow 119991, Russia.
| |
Collapse
|
38
|
Taubenberger AV, Girardo S, Träber N, Fischer-Friedrich E, Kräter M, Wagner K, Kurth T, Richter I, Haller B, Binner M, Hahn D, Freudenberg U, Werner C, Guck J. 3D Microenvironment Stiffness Regulates Tumor Spheroid Growth and Mechanics via p21 and ROCK. ACTA ACUST UNITED AC 2019; 3:e1900128. [PMID: 32648654 DOI: 10.1002/adbi.201900128] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Indexed: 01/01/2023]
Abstract
The mechanical properties of cancer cells and their microenvironment contribute to breast cancer progression. While mechanosensing has been extensively studied using 2D substrates, much less is known about it in a physiologically more relevant 3D context. Here it is demonstrated that breast cancer tumor spheroids, growing in 3D polyethylene glycol-heparin hydrogels, are sensitive to their environment stiffness. During tumor spheroid growth, compressive stresses of up to 2 kPa build up, as quantitated using elastic polymer beads as stress sensors. Atomic force microscopy reveals that tumor spheroid stiffness increases with hydrogel stiffness. Also, constituent cell stiffness increases in a Rho associated kinase (ROCK)- and F-actin-dependent manner. Increased hydrogel stiffness correlated with attenuated tumor spheroid growth, a higher proportion of cells in G0/G1 phase, and elevated levels of the cyclin-dependent kinase inhibitor p21. Drug-mediated ROCK inhibition not only reverses cell stiffening upon culture in stiff hydrogels but also increases tumor spheroid growth. Taken together, a mechanism by which the growth of a tumor spheroid can be regulated via cytoskeleton rearrangements in response to its mechanoenvironment is revealed here. Thus, the findings contribute to a better understanding of how cancer cells react to compressive stress when growing under confinement in stiff environments.
Collapse
Affiliation(s)
- Anna V Taubenberger
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Salvatore Girardo
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Max Planck Institute for the Science of Light, Max-Planck-Zentrum für Physik und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| | - Nicole Träber
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Elisabeth Fischer-Friedrich
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Martin Kräter
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Max Planck Institute for the Science of Light, Max-Planck-Zentrum für Physik und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| | - Katrin Wagner
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Thomas Kurth
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Isabel Richter
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Barbara Haller
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany
| | - Marcus Binner
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Dominik Hahn
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Carsten Werner
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Leibniz Institute of Polymer Research Dresden, Max Bergmann Center, Hohe Str. 6, 01069, Dresden, Germany
| | - Jochen Guck
- TU Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Fetscherstr. 105, 01307, Dresden, Germany.,Max Planck Institute for the Science of Light, Max-Planck-Zentrum für Physik und Medizin, Staudtstr. 2, 91058, Erlangen, Germany
| |
Collapse
|
39
|
Viji Babu PK, Radmacher M. Mechanics of Brain Tissues Studied by Atomic Force Microscopy: A Perspective. Front Neurosci 2019; 13:600. [PMID: 31258462 PMCID: PMC6587663 DOI: 10.3389/fnins.2019.00600] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/27/2019] [Indexed: 01/17/2023] Open
Abstract
Tissue morphology and mechanics are crucial to the regulation of organ function. Investigating the exceptionally complex tissue of the brain at the sub-micron scale is challenging due to the complex structure and softness of this tissue, despite the large interest of biologists, medical engineers, biophysicists, and others in this topic. Atomic force microscopy (AFM) both as an imaging and as a mechanical tool provides an excellent opportunity to study soft biological samples such as live brain tissues. Here we review the principles of AFM, the performance of AFM in tissue imaging and mechanical mapping of cells and tissues, and finally opening the prospects and challenges of probing the biophysical properties of brain tissue using AFM.
Collapse
|
40
|
Lekka M, Pabijan J, Orzechowska B. Morphological and mechanical stability of bladder cancer cells in response to substrate rigidity. Biochim Biophys Acta Gen Subj 2019; 1863:1006-1014. [DOI: 10.1016/j.bbagen.2019.03.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/14/2019] [Accepted: 03/11/2019] [Indexed: 01/13/2023]
|
41
|
Mollaeian K, Liu Y, Bi S, Wang Y, Ren J, Lu M. Nonlinear Cellular Mechanical Behavior Adaptation to Substrate Mechanics Identified by Atomic Force Microscope. Int J Mol Sci 2018; 19:ijms19113461. [PMID: 30400365 PMCID: PMC6274799 DOI: 10.3390/ijms19113461] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 10/13/2018] [Accepted: 10/31/2018] [Indexed: 12/15/2022] Open
Abstract
Cell–substrate interaction plays an important role in intracellular behavior and function. Adherent cell mechanics is directly regulated by the substrate mechanics. However, previous studies on the effect of substrate mechanics only focused on the stiffness relation between the substrate and the cells, and how the substrate stiffness affects the time-scale and length-scale of the cell mechanics has not yet been studied. The absence of this information directly limits the in-depth understanding of the cellular mechanotransduction process. In this study, the effect of substrate mechanics on the nonlinear biomechanical behavior of living cells was investigated using indentation-based atomic force microscopy. The mechanical properties and their nonlinearities of the cells cultured on four substrates with distinct mechanical properties were thoroughly investigated. Furthermore, the actin filament (F-actin) cytoskeleton of the cells was fluorescently stained to investigate the adaptation of F-actin cytoskeleton structure to the substrate mechanics. It was found that living cells sense and adapt to substrate mechanics: the cellular Young’s modulus, shear modulus, apparent viscosity, and their nonlinearities (mechanical property vs. measurement depth relation) were adapted to the substrates’ nonlinear mechanics. Moreover, the positive correlation between the cellular poroelasticity and the indentation remained the same regardless of the substrate stiffness nonlinearity, but was indeed more pronounced for the cells seeded on the softer substrates. Comparison of the F-actin cytoskeleton morphology confirmed that the substrate affects the cell mechanics by regulating the intracellular structure.
Collapse
Affiliation(s)
- Keyvan Mollaeian
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Yi Liu
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Siyu Bi
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Yifei Wang
- Department of Electrical and Computer Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Juan Ren
- Department of Mechanical Engineering, Iowa State University, Ames, IA 50011, USA.
| | - Meng Lu
- Department of Electrical and Computer Engineering, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
42
|
Muzzio NE, Pasquale MA, Marmisollé WA, von Bilderling C, Cortez ML, Pietrasanta LI, Azzaroni O. Self-assembled phosphate-polyamine networks as biocompatible supramolecular platforms to modulate cell adhesion. Biomater Sci 2018; 6:2230-2247. [PMID: 29978861 DOI: 10.1039/c8bm00265g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The modulation of cell adhesion via biologically inspired materials plays a key role in the development of realistic platforms to envisage not only mechanistic descriptions of many physiological and pathological processes but also new biointerfacial designs compatible with the requirements of biomedical devices. In this work, we show that the cell adhesion and proliferation of three different cell lines can be easily manipulated by using a novel biologically inspired supramolecular coating generated via dip coating of the working substrates in an aqueous solution of polyallylamine in the presence of phosphate anions-a simple one-step modification procedure. Our results reveal that selective cell adhesion can be controlled by varying the deposition time of the coating. Cell proliferation experiments showed a cell type-dependent quasi-exponential growth demonstrating the nontoxic properties of the supramolecular platform. After reaching a certain surface coverage, the supramolecular films based on phosphate-polyamine networks displayed antiadhesive activity towards cells, irrespective of the cell type. However and most interestingly, these antiadherent substrates developed strong adhesive properties after thermal annealing at 37 °C for 3 days. These results were interpreted based on the changes in the coating hydrophilicity, topography and stiffness, with the latter being assessed by atomic force microscopy imaging and indentation experiments. The reported approach is simple, robust and flexible, and would offer opportunities for the development of tunable, biocompatible interfacial architectures to control cell attachment for various biomedical applications.
Collapse
Affiliation(s)
- Nicolás E Muzzio
- Instituto de Investigaciones Fisicoquímicas Teóricas y Aplicadas (INIFTA), (UNLP, CONICET), Sucursal 4, Casilla de Correo 16, 1900 La Plata, Argentina.
| | | | | | | | | | | | | |
Collapse
|
43
|
Fabrication of hydroxyapatite/hydrophilic graphene composites and their modulation to cell behavior toward bone reconstruction engineering. Colloids Surf B Biointerfaces 2018; 173:512-520. [PMID: 30340179 DOI: 10.1016/j.colsurfb.2018.10.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/06/2018] [Accepted: 10/09/2018] [Indexed: 01/23/2023]
Abstract
Cell adhesion was the first step of bone reconstruction. While hydroxyapatite (HA)/graphene composites had been utilized for improving the cell adhesion and bone osteogenesis, the impact of cell adhesion and HA/graphene composites, especially HA/hydrophilic graphene (HG) composites, on internal interaction force and external surface properties remained poorly understood. Here, higher stability HA/HG composites were synthesized without extra ion introduction with in situ self-assembling method. And with XRD, FT-IR, XPS and Raman analyses, the evidences of the formation of HA and the introduction of HG was clear. TEM and SEM images showed the net-like spatial structure due to the internal interaction force between HA and HG, which provided the strain stimulation for cell adhesion. Subsequently, the external surface properties of HA/HG composites demonstrated that the roughness and hydrophilic ability of HA/HG composites could be artificially regulated by increasing the content of HG. Besides, the cell proliferation rate of HA/HG composites had been investigated. Compared to the intrinsic HA, HA/5%HG possessed the higher cell proliferation rate (264.81%) and promoted the spreading and growth of MC3T3-E1 cells. Finally, the regulation mechanism between HA/HG and cell adhesion were illuminated in detail. The excellent regular behavior of HA/HG composites for cell adhesion made them promising candidates for bone reconstruction and repairing. The present work provided the reference for the design of modifiable biomaterials and offered much inspiration for the future research of bone reconstruction engineering.
Collapse
|
44
|
Rianna C, Kumar P, Radmacher M. The role of the microenvironment in the biophysics of cancer. Semin Cell Dev Biol 2017; 73:107-114. [PMID: 28746843 DOI: 10.1016/j.semcdb.2017.07.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 01/23/2023]
Abstract
During the last decades, cell mechanics has been recognized as a quantitative measure to discriminate between many physiological and pathological states of single cells. In the field of biophysics of cancer, a large body of research has been focused on the comparison between normal and cancer mechanics and slowly the hypothesis that cancer cells are softer than their normal counterparts has been accepted, even though in situ tumor tissue is usually stiffer than the surrounding normal tissue. This corroborates the idea that the extra-cellular matrix (ECM) has a critical role in regulating tumor cell properties and behavior. Rearrangements in ECM can lead to changes in cancer cell mechanics and in specific conditions the general assumption about cancer cell softening could be confuted. Here, we highlight the contribution of ECM in cancer cell mechanics and argue that the statement that cancer cells are softer than normal cells should be firmly related to the properties of cell environment and the specific stage of cancer cell progression. In particular, we will discuss that when employing cell mechanics in cancer diagnosis and discrimination, the chemical, the topographical and - last but not least - the mechanical properties of the microenvironment are very important.
Collapse
Affiliation(s)
- Carmela Rianna
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany
| | - Prem Kumar
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany
| | - Manfred Radmacher
- Institute of Biophysics, University of Bremen, Otto-Hahn Allee 1, D-28359 Bremen, Germany.
| |
Collapse
|