101
|
Lee H, Bonin K, Guthold M. Human mammary epithelial cells in a mature, stratified epithelial layer flatten and stiffen compared to single and confluent cells. Biochim Biophys Acta Gen Subj 2021; 1865:129891. [PMID: 33689830 DOI: 10.1016/j.bbagen.2021.129891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 02/07/2021] [Accepted: 03/03/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND The epithelium forms a protective barrier against external biological, chemical and physical insults. So far, AFM-based, micro-mechanical measurements have only been performed on single cells and confluent cells, but not yet on cells in mature layers. METHODS Using a combination of atomic force, fluorescence and confocal microscopy, we determined the changes in stiffness, morphology and actin distribution of human mammary epithelial cells (HMECs) as they transition from single cells to confluency to a mature layer. RESULTS Single HMECs have a tall, round (planoconvex) morphology, have actin stress fibers at the base, have diffuse cortical actin, and have a stiffness of 1 kPa. Confluent HMECs start to become flatter, basal actin stress fibers start to disappear, and actin accumulates laterally where cells abut. Overall stiffness is still 1 kPa with two-fold higher stiffness in the abutting regions. As HMECs mature and form multilayered structures, cells on apical surfaces become flatter (apically more level), wider, and seven times stiffer (mean, 7 kPa) than single and confluent cells. The main drivers of these changes are actin filaments, as cells show strong actin accumulation in the regions where cells adjoin, and in the apical regions. CONCLUSIONS HMECs stiffen, flatten and redistribute actin upon transiting from single cells to mature, confluent layers. GENERAL SIGNIFICANCE Our findings advance the understanding of breast ductal morphogenesis and mechanical homeostasis.
Collapse
Affiliation(s)
- Hyunsu Lee
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Keith Bonin
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Martin Guthold
- Department of Physics, Wake Forest University, Winston-Salem, NC 27109, USA.
| |
Collapse
|
102
|
Graybill PM, Bollineni RK, Sheng Z, Davalos RV, Mirzaeifar R. A constriction channel analysis of astrocytoma stiffness and disease progression. BIOMICROFLUIDICS 2021; 15:024103. [PMID: 33763160 PMCID: PMC7968935 DOI: 10.1063/5.0040283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/23/2021] [Indexed: 05/12/2023]
Abstract
Studies have demonstrated that cancer cells tend to have reduced stiffness (Young's modulus) compared to their healthy counterparts. The mechanical properties of primary brain cancer cells, however, have remained largely unstudied. To investigate whether the stiffness of primary brain cancer cells decreases as malignancy increases, we used a microfluidic constriction channel device to deform healthy astrocytes and astrocytoma cells of grade II, III, and IV and measured the entry time, transit time, and elongation. Calculating cell stiffness directly from the experimental measurements is not possible. To overcome this challenge, finite element simulations of the cell entry into the constriction channel were used to train a neural network to calculate the stiffness of the analyzed cells based on their experimentally measured diameter, entry time, and elongation in the channel. Our study provides the first calculation of stiffness for grades II and III astrocytoma and is the first to apply a neural network analysis to determine cell mechanical properties from a constriction channel device. Our results suggest that the stiffness of astrocytoma cells is not well-correlated with the cell grade. Furthermore, while other non-central-nervous-system cell types typically show reduced stiffness of malignant cells, we found that most astrocytoma cell lines had increased stiffness compared to healthy astrocytes, with lower-grade astrocytoma having higher stiffness values than grade IV glioblastoma. Differences in nucleus-to-cytoplasm ratio only partly explain differences in stiffness values. Although our study does have limitations, our results do not show a strong correlation of stiffness with cell grade, suggesting that other factors may play important roles in determining the invasive capability of astrocytoma. Future studies are warranted to further elucidate the mechanical properties of astrocytoma across various pathological grades.
Collapse
Affiliation(s)
| | - R. K. Bollineni
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Z. Sheng
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine and Virginia Tech Fralin Biomedical Research Institute, Roanoke, Virginia 24016, USA
| | - R. V. Davalos
- Authors to whom correspondence should be addressed: and
| | - R. Mirzaeifar
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
103
|
Li M, Xi N, Wang YC, Liu LQ. Atomic force microscopy for revealing micro/nanoscale mechanics in tumor metastasis: from single cells to microenvironmental cues. Acta Pharmacol Sin 2021; 42:323-339. [PMID: 32807839 PMCID: PMC8027022 DOI: 10.1038/s41401-020-0494-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023]
Abstract
Mechanics are intrinsic properties which appears throughout the formation, development, and aging processes of biological systems. Mechanics have been shown to play important roles in regulating the development and metastasis of tumors, and understanding tumor mechanics has emerged as a promising way to reveal the underlying mechanisms guiding tumor behaviors. In particular, tumors are highly complex diseases associated with multifaceted factors, including alterations in cancerous cells, tissues, and organs as well as microenvironmental cues, indicating that investigating tumor mechanics on multiple levels is significantly helpful for comprehensively understanding the effects of mechanics on tumor progression. Recently, diverse techniques have been developed for probing the mechanics of tumors, among which atomic force microscopy (AFM) has appeared as an excellent platform enabling simultaneously characterizing the structures and mechanical properties of living biological systems ranging from individual molecules and cells to tissue samples with unprecedented spatiotemporal resolution, offering novel possibilities for understanding tumor physics and contributing much to the studies of cancer. In this review, we survey the recent progress that has been achieved with the use of AFM for revealing micro/nanoscale mechanics in tumor development and metastasis. Challenges and future progress are also discussed.
Collapse
Affiliation(s)
- Mi Li
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Ning Xi
- Department of Industrial and Manufacturing Systems Engineering, The University of Hong Kong, Hong Kong, China
| | - Yue-Chao Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lian-Qing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China.
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China.
- University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
104
|
Yasukuni R, Minamino D, Iino T, Araki T, Takao K, Yamada S, Bessho Y, Matsui T, Hosokawa Y. Pulsed laser activated impulse response encoder (PLAIRE): sensitive evaluation of surface cellular stiffness on zebrafish embryos. BIOMEDICAL OPTICS EXPRESS 2021; 12:1366-1374. [PMID: 33796359 PMCID: PMC7984775 DOI: 10.1364/boe.414338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 06/12/2023]
Abstract
Mechanical properties of cells and tissues closely link to their architectures and physiological functions. To obtain the mechanical information of submillimeter scale small biological objects, we recently focused on the object vibration responses when excited by a femtosecond laser-induced impulsive force. These responses are monitored by the motion of an AFM cantilever placed on top of a sample. In this paper, we examined the surface cellular stiffness of zebrafish embryos based on excited vibration forms in different cytoskeletal states. The vibration responses were more sensitive to their surface cellular stiffness in comparison to the Young's modulus obtained by a conventional AFM force curve measurement.
Collapse
Affiliation(s)
- Ryohei Yasukuni
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Daiki Minamino
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takanori Iino
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takashi Araki
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Kohei Takao
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Sohei Yamada
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Yasumasa Bessho
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Takaaki Matsui
- Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| | - Yoichiroh Hosokawa
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara 630-0192, Japan
| |
Collapse
|
105
|
Hope JM, Bersi MR, Dombroski JA, Clinch AB, Pereles RS, Merryman WD, King MR. Circulating prostate cancer cells have differential resistance to fluid shear stress-induced cell death. J Cell Sci 2021; 134:jcs.251470. [PMID: 33526716 PMCID: PMC7929932 DOI: 10.1242/jcs.251470] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Circulating tumor cells (CTCs) are exposed to fluid shear stress (FSS) of greater than 1000 dyn/cm2 (100 Pa) in circulation. Normally, CTCs that are exposed to FSS of this magnitude die. However, some CTCs develop resistance to this FSS, allowing them to colonize distant organs. We explored how prostate CTCs can resist cell death in response to forces of this magnitude. The DU145, PC3 and LNCaP human prostate cancer cell lines were used to represent cells of different metastatic origins. The cell lines were briefly treated with an average FSS of 3950 dyn/cm2 (395 Pa) using a 30 G needle and a syringe pump. DU145 cells had no change in cell viability, PC3 cells had some cell death and LNCaP cells exhibited significant cell death. These cell death responses correlated with increased cell membrane damage, less efficient membrane repair and increased stiffness. Additionally, FSS treatment prevented the LNCaP FSS-sensitive cell line from forming a growing tumor in vivo. This suggests that these properties play a role in FSS resistance and could represent potential targets for disrupting blood-borne metastasis. Summary: Prostate cancer cells have different sensitivities to fluid forces that alter their resistance to elevated blood flow-level fluid shear stress.
Collapse
Affiliation(s)
- Jacob M Hope
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Matthew R Bersi
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Jenna A Dombroski
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Andrea B Clinch
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Rebecca S Pereles
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA
| |
Collapse
|
106
|
Neurotoxic Effect of Flavonol Myricetin in the Presence of Excess Copper. Molecules 2021; 26:molecules26040845. [PMID: 33562817 PMCID: PMC7914656 DOI: 10.3390/molecules26040845] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 11/16/2022] Open
Abstract
Oxidative stress (OS) induced by the disturbed homeostasis of metal ions is one of the pivotal factors contributing to neurodegeneration. The aim of the present study was to investigate the effects of flavonoid myricetin on copper-induced toxicity in neuroblastoma SH-SY5Y cells. As determined by the MTT method, trypan blue exclusion assay and measurement of ATP production, myricetin heightened the toxic effects of copper and exacerbated cell death. It also increased copper-induced generation of reactive oxygen species, indicating the prooxidative nature of its action. Furthermore, myricetin provoked chromatin condensation and loss of membrane integrity without caspase-3 activation, suggesting the activation of both caspase-independent programmed cell death and necrosis. At the protein level, myricetin-induced upregulation of PARP-1 and decreased expression of Bcl-2, whereas copper-induced changes in the expression of p53, p73, Bax and NME1 were not further affected by myricetin. Inhibitors of ERK1/2 and JNK kinases, protein kinase A and L-type calcium channels exacerbated the toxic effects of myricetin, indicating the involvement of intracellular signaling pathways in cell death. We also employed atomic force microscopy (AFM) to evaluate the morphological and mechanical properties of SH-SY5Y cells at the nanoscale. Consistent with the cellular and molecular methods, this biophysical approach also revealed a myricetin-induced increase in cell surface roughness and reduced elasticity. Taken together, we demonstrated the adverse effects of myricetin, pointing out that caution is required when considering powerful antioxidants for adjuvant therapy in copper-related neurodegeneration.
Collapse
|
107
|
Kar S, Katti DR, Katti KS. Evaluation of quasi-static and dynamic nanomechanical properties of bone-metastatic breast cancer cells using a nanoclay cancer testbed. Sci Rep 2021; 11:3096. [PMID: 33542384 PMCID: PMC7862348 DOI: 10.1038/s41598-021-82664-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/20/2021] [Indexed: 01/07/2023] Open
Abstract
In recent years, there has been increasing interest in investigating the mechanical properties of individual cells to delineate disease mechanisms. Reorganization of cytoskeleton facilitates the colonization of metastatic breast cancer at bone marrow space, leading to bone metastasis. Here, we report evaluation of mechanical properties of two breast cancer cells with different metastatic ability at the site of bone metastases, using quasi-static and dynamic nanoindentation methods. Our results showed that the significant reduction in elastic modulus along with increased liquid-like behavior of bone metastasized MCF-7 cells was induced by depolymerization and reorganization of F-actin to the adherens junctions, whereas bone metastasized MDA-MB-231 cells showed insignificant changes in elastic modulus and F-actin reorganization over time, compared to their respective as-received counterparts. Taken together, our data demonstrate evolution of breast cancer cell mechanics at bone metastases.
Collapse
Affiliation(s)
- Sumanta Kar
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering, North Dakota State University, Fargo, ND, 58108, USA
| | - Dinesh R Katti
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering, North Dakota State University, Fargo, ND, 58108, USA
| | - Kalpana S Katti
- Center for Engineered Cancer Test Beds, Department of Civil and Environmental Engineering, North Dakota State University, Fargo, ND, 58108, USA.
| |
Collapse
|
108
|
Richter S, Martin R, Gutzeit HO, Knölker HJ. In vitro and in vivo effects of inhibitors on actin and myosin. Bioorg Med Chem 2021; 30:115928. [PMID: 33341499 DOI: 10.1016/j.bmc.2020.115928] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 11/18/2022]
Abstract
The interaction of actin and myosin is essential for cell migration. We have identified kaempferol and pentahalogenated pseudilins as efficient inhibitors of migration of MDA-MB-231 breast adenocarcinoma cells. The compounds were studied with respect to possible effects on myosin-2-ATPase activity. The pentahalogenated pseudilins inhibited the enzyme activity in vitro. Flavonoids showed no effect on enzyme activity. The polymerization dynamics of actin was measured to test whether the integrity of F-actin is essential for the migration of MDA-MB-231 cells. Quercetin and kaempferol depolymerized F-actin with similar efficiencies as found for the pentahalogenated pseudilins, whereas epigallocatechin showed the weakest effect. As the inhibitory effect on cell migration may be caused by a toxic effect, we have performed a cytotoxicity test and, furthermore, investigated the influence of the test compounds on cardiac function in eleutheroembryos of medaka (Oryzias latipes). Compared with the pentahalogenated pseudilins, the cytotoxic and cardiotoxic effects of flavonoids on medaka embryos were found to be moderate.
Collapse
Affiliation(s)
- Sabine Richter
- Faculty of Biology, TU Dresden, Zellescher Weg 20b, 01217 Dresden, Germany
| | - René Martin
- Faculty of Chemistry, TU Dresden, Bergstrasse 66, 01069 Dresden, Germany
| | - Herwig O Gutzeit
- Faculty of Biology, TU Dresden, Zellescher Weg 20b, 01217 Dresden, Germany.
| | | |
Collapse
|
109
|
Romanov V, Silvani G, Zhu H, Cox CD, Martinac B. An Acoustic Platform for Single-Cell, High-Throughput Measurements of the Viscoelastic Properties of Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2005759. [PMID: 33326190 DOI: 10.1002/smll.202005759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/01/2020] [Indexed: 06/12/2023]
Abstract
Cellular processes including adhesion, migration, and differentiation are governed by the distinct mechanical properties of each cell. Importantly, the mechanical properties of individual cells can vary depending on local physical and biochemical cues in a time-dependent manner resulting in significant inter-cell heterogeneity. While several different methods have been developed to interrogate the mechanical properties of single cells, throughput to capture this heterogeneity remains an issue. Here, single-cell, high-throughput characterization of adherent cells is demonstrated using acoustic force spectroscopy (AFS). AFS works by simultaneously, acoustically driving tens to hundreds of silica beads attached to cells away from the cell surface, allowing the user to measure the stiffness of adherent cells under multiple experimental conditions. It is shown that cells undergo marked changes in viscoelasticity as a function of temperature, by altering the temperature within the AFS microfluidic circuit between 21 and 37 °C. In addition, quantitative differences in cells exposed to different pharmacological treatments specifically targeting the membrane-cytoskeleton interface are shown. Further, the high-throughput format of the AFS is utilized to rapidly probe, in excess of 1000 cells, three different cell lines expressing different levels of a mechanosensitive protein, Piezo1, demonstrating the ability to differentiate between cells based on protein expression levels.
Collapse
Affiliation(s)
- Valentin Romanov
- Department of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Giulia Silvani
- Department of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, NSW, 2010, Australia
| | - Huiyu Zhu
- Faculty of Science, University of Technology Sydney, Ultimo, Sydney, NSW, 2007, Australia
| | - Charles D Cox
- Department of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
| | - Boris Martinac
- Department of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2010, Australia
| |
Collapse
|
110
|
Gryadunova AA, Koudan EV, Rodionov SA, Pereira FDAS, Meteleva NY, Kasyanov VA, Parfenov VA, Kovalev AV, Khesuani YD, Mironov VA, Bulanova EA. Cytoskeleton systems contribute differently to the functional intrinsic properties of chondrospheres. Acta Biomater 2020; 118:141-152. [PMID: 33045401 DOI: 10.1016/j.actbio.2020.10.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
Cytoskeleton systems, actin microfilaments, microtubules (MTs) and intermediate filaments (IFs) provide the biomechanical stability and spatial organization in cells. To understand the specific contributions of each cytoskeleton systems to intrinsic properties of spheroids, we've scrutinized the effects of the cytoskeleton perturbants, cytochalasin D (Cyto D), nocodazole (Noc) and withaferin A (WFA) on fusion, spreading on adhesive surface, morphology and biomechanics of chondrospheres (CSs). We confirmed that treatment with Cyto D but not with Noc or WFA severely affected CSs fusion and spreading dynamics and significantly reduced biomechanical properties of cell aggregates. Noc treatment affected spheroids spreading but not the fusion and surprisingly enhanced their stiffness. Vimentin intermediate filaments (VIFs) reorganization affected CSs spreading only. The analysis of all three cytoskeleton systems contribution to spheroids intrinsic properties was performed for the first time.
Collapse
Affiliation(s)
- Anna A Gryadunova
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation; Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russian Federation.
| | - Elizaveta V Koudan
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation.
| | - Sergey A Rodionov
- N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, Moscow 127299, Russian Federation
| | - F D A S Pereira
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation
| | - Nina Yu Meteleva
- I.D. Papanin Institute for Biology of Inland Waters RAS, Borok 152742, Russian Federation
| | - Vladimir A Kasyanov
- Riga Stradins University, Riga LV-1007, Latvia; Riga Technical University, Riga LV-1658, Latvia
| | - Vladislav A Parfenov
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation
| | - Alexey V Kovalev
- N.N. Priorov National Medical Research Center of Traumatology and Orthopedics, Moscow 127299, Russian Federation
| | - Yusef D Khesuani
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation
| | - Vladimir A Mironov
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation; Institute for Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russian Federation
| | - Elena A Bulanova
- Laboratory for Biotechnological Research 3D Bioprinting Solutions, Moscow 115409, Russian Federation.
| |
Collapse
|
111
|
Kubiak A, Zieliński T, Pabijan J, Lekka M. Nanomechanics in Monitoring the Effectiveness of Drugs Targeting the Cancer Cell Cytoskeleton. Int J Mol Sci 2020; 21:E8786. [PMID: 33233645 PMCID: PMC7699791 DOI: 10.3390/ijms21228786] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/19/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Increasing attention is devoted to the use of nanomechanics as a marker of various pathologies. Atomic force microscopy (AFM) is one of the techniques that could be applied to quantify the nanomechanical properties of living cells with a high spatial resolution. Thus, AFM offers the possibility to trace changes in the reorganization of the cytoskeleton in living cells. Impairments in the structure, organization, and functioning of two main cytoskeletal components, namely, actin filaments and microtubules, cause severe effects, leading to cell death. That is why these cytoskeletal components are targets for antitumor therapy. This review intends to describe the gathered knowledge on the capability of AFM to trace the alterations in the nanomechanical properties of living cells induced by the action of antitumor drugs that could translate into their effectiveness.
Collapse
Affiliation(s)
| | | | | | - Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Kraków, Poland; (A.K.); (T.Z.); (J.P.)
| |
Collapse
|
112
|
Weber A, Zbiral B, Iturri J, Benitez R, Toca-Herrera JL. Measuring (biological) materials mechanics with atomic force microscopy. 2. Influence of the loading rate and applied force (colloidal particles). Microsc Res Tech 2020; 84:1078-1088. [PMID: 33179834 DOI: 10.1002/jemt.23643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/25/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022]
Abstract
Atomic force microscopy (AFM) is the most often used tool to study the mechanical properties of eukaryotic cells. Due to their complex assembly, cells show viscoelastic properties. When performing experiments, one has to consider the influence of both loading rate and maximum load on the measured mechanical properties. Here, we employed colloidal particles of various sizes (from 2 to 20 μm diameter) to perform force spectroscopy measurements on endothelial cells at loading rates varying from 0.1 to 50 μm/s, and maximum loads ranging from 1 to 25 nN. We were able to determine the non-linear dependence of cell viscoelastic properties on the loading rate which followed a weak power law. In addition, we show that previous loading at high forces leads to a stiffening of cells. Based on these results we discuss a road map for determining cell mechanical properties using AFM. Finally, this work provides an experimental framework for cell mechanical measurements using force-cycle experiments.
Collapse
Affiliation(s)
- Andreas Weber
- Department of Nanobiotechnology, Institute for Biophysics, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Barbara Zbiral
- Department of Nanobiotechnology, Institute for Biophysics, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Jagoba Iturri
- Department of Nanobiotechnology, Institute for Biophysics, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| | - Rafael Benitez
- Departamento de Matemáticas para la Economía y la Empresa, Facultad de Economía, Universidad de Valencia, Valencia, Spain
| | - José L Toca-Herrera
- Department of Nanobiotechnology, Institute for Biophysics, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| |
Collapse
|
113
|
Liu X, Wei Y, Li W, Li B, Liu L. Cytoskeleton induced the changes of microvilli and mechanical properties in living cells by atomic force microscopy. J Cell Physiol 2020; 236:3725-3733. [PMID: 33169846 DOI: 10.1002/jcp.30110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 01/05/2023]
Abstract
The cytoskeleton acts as a scaffold for membrane protrusion, such as microvilli. However, the relationship between the characteristics of microvilli and cytoskeleton remains poorly understood under the physiological state. To investigate the role of the cytoskeleton in regulating microvilli and cellular mechanical properties, atomic force microscopy (AFM) was used to detect the dynamic characteristics of microvillus morphology and elastic modulus of living HeLa cells. First, HeLa and MCF-7 cell lines were stained with Fluor-488-phalloidin and microtubules antibody. Then, the microvilli morphology was analyzed by high-resolution images of AFM in situ. Furthermore, changes in elastic modulus were investigated by the force curve of AFM. Fluorescence microscopy and AFM results revealed that destroyed microfilaments led to a smaller microvilli size, whereas the increase in the aggregation and number of microfilaments led to a larger microvilli size. The destruction and aggregation of microfilaments remarkably affected the mechanical properties of HeLa cells. Microtubule-related drugs induced the change of microtubule, but we failed to note significant differences in microvilli morphology and mechanical properties of cells. In summary, our results unraveled the relationship between microfilaments and the structure of microvilli and Young's modulus in living HeLa cells, which would contribute to the further understanding of the physiological function of the cytoskeleton in vivo.
Collapse
Affiliation(s)
- Xueyan Liu
- Key Laboratory of Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education of China, Wenzhou Medical University, Wenzhou, China
| | - Yuhui Wei
- Division of Physical Biology and Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Chinese Academy of Sciences, Shanghai Institute of Applied Physics, Shanghai, China.,Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Wei Li
- Key Laboratory of Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education of China, Wenzhou Medical University, Wenzhou, China.,Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, China
| | - Bin Li
- Division of Physical Biology and Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Chinese Academy of Sciences, Shanghai Institute of Applied Physics, Shanghai, China.,Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Lin Liu
- Key Laboratory of Medicine, School of Laboratory Medicine and Life Sciences, Ministry of Education of China, Wenzhou Medical University, Wenzhou, China.,Division of Physical Biology and Bioimaging Centre, Shanghai Synchrotron Radiation Facility, CAS Key Laboratory of Interfacial Physics and Technology, Chinese Academy of Sciences, Shanghai Institute of Applied Physics, Shanghai, China.,Zhejiang Provincial Key Laboratory of Medical Genetics, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
114
|
Marcksl1 modulates endothelial cell mechanoresponse to haemodynamic forces to control blood vessel shape and size. Nat Commun 2020; 11:5476. [PMID: 33127887 PMCID: PMC7603353 DOI: 10.1038/s41467-020-19308-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 10/02/2020] [Indexed: 11/16/2022] Open
Abstract
The formation of vascular tubes is driven by extensive changes in endothelial cell (EC) shape. Here, we have identified a role of the actin-binding protein, Marcksl1, in modulating the mechanical properties of EC cortex to regulate cell shape and vessel structure during angiogenesis. Increasing and depleting Marcksl1 expression level in vivo results in an increase and decrease, respectively, in EC size and the diameter of microvessels. Furthermore, endothelial overexpression of Marcksl1 induces ectopic blebbing on both apical and basal membranes, during and after lumen formation, that is suppressed by reduced blood flow. High resolution imaging reveals that Marcksl1 promotes the formation of linear actin bundles and decreases actin density at the EC cortex. Our findings demonstrate that a balanced network of linear and branched actin at the EC cortex is essential in conferring cortical integrity to resist the deforming forces of blood flow to regulate vessel structure. During lumen formation in blood vessels, endothelial cells become exposed to hemodynamic forces that induce membrane blebbing and changes in cell shape. Here, the authors show endothelial cells develop an actin-based protective mechanism in the cell cortex that prevents excessive blebbing to control cell shape and vessel diameter.
Collapse
|
115
|
Visuo-haptic virtual exploration of single cell morphology and mechanics based on AFM mapping in fast mode. JOURNAL OF MICRO-BIO ROBOTICS 2020. [DOI: 10.1007/s12213-020-00140-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
116
|
Lüchtefeld I, Bartolozzi A, Mejía Morales J, Dobre O, Basso M, Zambelli T, Vassalli M. Elasticity spectra as a tool to investigate actin cortex mechanics. J Nanobiotechnology 2020; 18:147. [PMID: 33081777 PMCID: PMC7576730 DOI: 10.1186/s12951-020-00706-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/09/2020] [Indexed: 12/24/2022] Open
Abstract
Background The mechanical properties of single living cells have proven to be a powerful marker of the cell physiological state. The use of nanoindentation-based single cell force spectroscopy provided a wealth of information on the elasticity of cells, which is still largely to be exploited. The simplest model to describe cell mechanics is to treat them as a homogeneous elastic material and describe it in terms of the Young’s modulus. Beside its simplicity, this approach proved to be extremely informative, allowing to assess the potential of this physical indicator towards high throughput phenotyping in diagnostic and prognostic applications. Results Here we propose an extension of this analysis to explicitly account for the properties of the actin cortex. We present a method, the Elasticity Spectra, to calculate the apparent stiffness of the cell as a function of the indentation depth and we suggest a simple phenomenological approach to measure the thickness and stiffness of the actin cortex, in addition to the standard Young’s modulus. Conclusions The Elasticity Spectra approach is tested and validated on a set of cells treated with cytoskeleton-affecting drugs, showing the potential to extend the current representation of cell mechanics, without introducing a detailed and complex description of the intracellular structure.![]()
Collapse
Affiliation(s)
- Ines Lüchtefeld
- Laboratory of Biosensors and Bioelectronics, ETH Zürich, Gloriastrasse 35, 8092, Zürich, Switzerland
| | - Alice Bartolozzi
- Dipartimento di Ingegneria dell'Informazione, Università degli studi di Firenze, Via di S. Marta 3, 50139, Firenze, Italy
| | - Julián Mejía Morales
- Institut de Physique de Nice, Université Côte d'Azur, 1361 Route des Lucioles, 06560, Valbonne, France.,Dipartimento di Medicina Sperimentale, Università degli studi di Genova, Via Leon Battista Alberti 2, 16132, Genova, Italy
| | - Oana Dobre
- James Watt School of Engineering, University of Glasgow, Oakfield avenue, Glasgow, G12 8LT, UK
| | - Michele Basso
- Dipartimento di Ingegneria dell'Informazione, Università degli studi di Firenze, Via di S. Marta 3, 50139, Firenze, Italy
| | - Tomaso Zambelli
- Laboratory of Biosensors and Bioelectronics, ETH Zürich, Gloriastrasse 35, 8092, Zürich, Switzerland
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Oakfield avenue, Glasgow, G12 8LT, UK.
| |
Collapse
|
117
|
Ombid RJL, Oyong GG, Cabrera EC, Espulgar WV, Saito M, Tamiya E, Pobre RF. In-vitro study of monocytic THP-1 leukemia cell membrane elasticity with a single-cell microfluidic-assisted optical trapping system. BIOMEDICAL OPTICS EXPRESS 2020; 11:6027-6037. [PMID: 33150003 PMCID: PMC7587289 DOI: 10.1364/boe.402526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/20/2020] [Accepted: 09/23/2020] [Indexed: 06/11/2023]
Abstract
We studied the elastic profile of monocytic THP-1 leukemia cells using a microfluidic-assisted optical trap. A 2-µm fused silica bead was optically trapped to mechanically dent an immobilized single THP-1 monocyte sieved on a 15-µm microfluidic capture chamber. Cells treated with Zeocin and untreated cells underwent RT-qPCR analysis to determine cell apoptosis through gene expression in relation to each cell's deformation profile. Results showed that untreated cells with 43.05 ± 6.68 Pa are more elastic compared to the treated cells with 15.81 ± 2.94 Pa. THP-1 monocyte's elastic modulus is indicative of cell apoptosis shown by upregulated genes after Zeocin treatment. This study clearly showed that the developed technique can be used to distinguish between cells undergoing apoptosis and cells not undergoing apoptosis and which may apply to the study of other cells and other cell states as well.
Collapse
Affiliation(s)
- Ric John L. Ombid
- OPTICS Research Unit, CENSER, De La Salle University (DLSU), Manila, Philippines
- Optics and Instrumentation Physics Laboratory, Physics Department, DLSU, Manila, Philippines
| | - Glenn G. Oyong
- OPTICS Research Unit, CENSER, De La Salle University (DLSU), Manila, Philippines
- Molecular Science Unit Laboratory, CENSER, DLSU, Manila, Philippines
| | - Esperanza C. Cabrera
- Biology Department, DLSU, Manila, Philippines
- Molecular Science Unit Laboratory, CENSER, DLSU, Manila, Philippines
| | - Wilfred V. Espulgar
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Japan
| | - Masato Saito
- Department of Applied Physics, Graduate School of Engineering, Osaka University, Japan
- Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Photonics Center, Osaka University, Osaka 565-0871, Japan
| | - Eiichi Tamiya
- Advanced Photonics and Biosensing Open Innovation Laboratory, AIST-Osaka University, Photonics Center, Osaka University, Osaka 565-0871, Japan
- The Institute of Scientific and Industrial Research, Osaka University, Japan
| | - Romeric F. Pobre
- OPTICS Research Unit, CENSER, De La Salle University (DLSU), Manila, Philippines
- Optics and Instrumentation Physics Laboratory, Physics Department, DLSU, Manila, Philippines
| |
Collapse
|
118
|
Viscoelasticity and Volume of Cortical Neurons under Glutamate Excitotoxicity and Osmotic Challenges. Biophys J 2020; 119:1712-1723. [PMID: 33086042 DOI: 10.1016/j.bpj.2020.09.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/28/2020] [Accepted: 09/21/2020] [Indexed: 01/15/2023] Open
Abstract
Neural activity depends on the maintenance of ionic and osmotic homeostasis. Under these conditions, the cell volume must be regulated to maintain optimal neural function. A disturbance in the neuronal volume regulation often occurs in pathological conditions such as glutamate excitotoxicity. The cell volume, mechanical properties, and actin cytoskeleton structure are tightly connected to achieve the cell homeostasis. Here, we studied the effects of glutamate-induced excitotoxicity, external osmotic pressure, and inhibition of actin polymerization on the viscoelastic properties and volume of neurons. Atomic force microscopy was used to map the viscoelastic properties of neurons in time-series experiments to observe the dynamical changes and a possible recovery. The data obtained on cultured rat cortical neurons were compared with the data obtained on rat fibroblasts. The neurons were found to be more responsive to the osmotic challenges but less sensitive to the inhibition of actin polymerization than fibroblasts. The alterations of the viscoelastic properties caused by glutamate excitotoxicity were similar to those induced by the hypoosmotic stress, but, in contrast to the latter, they did not recover after the glutamate removal. These data were consistent with the dynamic volume changes estimated using ratiometric fluorescent dyes. The recovery after the glutamate-induced excitotoxicity was slow or absent because of a steady increase in intracellular calcium and sodium concentrations. The viscoelastic parameters and their changes were related to such parameters as the actin cortex stiffness, tension, and cytoplasmic viscosity.
Collapse
|
119
|
Nagayama K, Ohata S, Obata S, Sato A. Macroscopic and microscopic analysis of the mechanical properties and adhesion force of cells using a single cell tensile test and atomic force microscopy: Remarkable differences in cell types. J Mech Behav Biomed Mater 2020; 110:103935. [PMID: 32957229 DOI: 10.1016/j.jmbbm.2020.103935] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/16/2020] [Accepted: 06/13/2020] [Indexed: 01/11/2023]
Abstract
Many experimental techniques have been reported to provide knowledge of the mechanical behavior of cells from biomechanical viewpoints, however, it is unclear how the intercellular structural differences influence macroscopic and microscopic mechanical properties of cells. The aim of our study is to clarify the comprehensive mechanical properties and cell-substrate adhesion strength of cells, and the correlation with intracellular structure in different cell types. We developed an originally designed micro tensile tester, and performed a single cell tensile test to estimate whole cell tensile stiffness and adhesion strength of normal vascular smooth muscle cells (VSMCs) and cervical cancer HeLa cells: one half side of the specimen cell was lifted up by a glass microneedle, then stretched until the cell detached from the substrate, while force was simultaneously measured. The tensile stiffness and adhesion strength were 49 ± 10 nN/% and 870 ± 430 nN, respectively, in VSMCs (mean ± SD, n = 8), and 19 ± 17 nN/% and 320 ± 160 nN, respectively, in HeLa cells (n = 9). The difference was more definite in the surface elastic modulus map obtained by atomic force microscopy, indicating that the internal tension of the actin cytoskeleton was significantly higher in VSMCs than in HeLa cells. Structural analysis with confocal microscopy revealed that VSMCs had a significant alignment of F-actin cytoskeleton with mature focal adhesion, contrary to the randomly oriented F-actin with smaller focal adhesion of HeLa cells, indicating that structural arrangement of the actin cytoskeleton and their mechanical tension generated the differences in cell mechanical properties and adhesion forces. The finding strongly suggests that the mechanical and structural differences in each cell type are deeply involved with their physiological functions.
Collapse
Affiliation(s)
- Kazuaki Nagayama
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan.
| | - Shigeaki Ohata
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan
| | - Shota Obata
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan
| | - Akiko Sato
- Micro-Nano Biomechanics Laboratory, Department of Mechanical Systems Engineering, Ibaraki University, Nakanarusawa-cho, Hitachi, 316-8511, Japan
| |
Collapse
|
120
|
Guo A, Wang B, Lyu C, Li W, Wu Y, Zhu L, Bi R, Huang C, Li JJ, Du Y. Consistent apparent Young's modulus of human embryonic stem cells and derived cell types stabilized by substrate stiffness regulation promotes lineage specificity maintenance. CELL REGENERATION 2020; 9:15. [PMID: 32880028 PMCID: PMC7467757 DOI: 10.1186/s13619-020-00054-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/15/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND Apparent Young's modulus (AYM), which reflects the fundamental mechanical property of live cells measured by atomic force microscopy and is determined by substrate stiffness regulated cytoskeletal organization, has been investigated as potential indicators of cell fate in specific cell types. However, applying biophysical cues, such as modulating the substrate stiffness, to regulate AYM and thereby reflect and/or control stem cell lineage specificity for downstream applications, remains a primary challenge during in vitro stem cell expansion. Moreover, substrate stiffness could modulate cell heterogeneity in the single-cell stage and contribute to cell fate regulation, yet the indicative link between AYM and cell fate determination during in vitro dynamic cell expansion (from single-cell stage to multi-cell stage) has not been established. RESULTS Here, we show that the AYM of cells changed dynamically during passaging and proliferation on substrates with different stiffness. Moreover, the same change in substrate stiffness caused different patterns of AYM change in epithelial and mesenchymal cell types. Embryonic stem cells and their derived progenitor cells exhibited distinguishing AYM changes in response to different substrate stiffness that had significant effects on their maintenance of pluripotency and/or lineage-specific characteristics. On substrates that were too rigid or too soft, fluctuations in AYM occurred during cell passaging and proliferation that led to a loss in lineage specificity. On a substrate with 'optimal' stiffness (i.e., 3.5 kPa), the AYM was maintained at a constant level that was consistent with the parental cells during passaging and proliferation and led to preservation of lineage specificity. The effects of substrate stiffness on AYM and downstream cell fate were correlated with intracellular cytoskeletal organization and nuclear/cytoplasmic localization of YAP. CONCLUSIONS In summary, this study suggests that optimal substrate stiffness regulated consistent AYM during passaging and proliferation reflects and contributes to hESCs and their derived progenitor cells lineage specificity maintenance, through the underlying mechanistic pathways of stiffness-induced cytoskeletal organization and the downstream YAP signaling. These findings highlighted the potential of AYM as an indicator to select suitable substrate stiffness for stem cell specificity maintenance during in vitro expansion for regenerative applications.
Collapse
Affiliation(s)
- Anqi Guo
- Department of Biomedical Engineering, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, School of Medicine, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bingjie Wang
- Department of Biomedical Engineering, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, School of Medicine, Tsinghua University, Beijing, 100084, China.,School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Cheng Lyu
- Department of Biomedical Engineering, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Wenjing Li
- Department of Biomedical Engineering, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Yaozu Wu
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, 63130, USA
| | - Lu Zhu
- Institute of Systems Engineering, Academy of Military Sciences, Beijing, 100071, China
| | - Ran Bi
- Department of Biomedical Engineering, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Chenyu Huang
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Jiao Jiao Li
- Kolling Institute, University of Sydney, Sydney, NSW, 2006, Australia
| | - Yanan Du
- Department of Biomedical Engineering, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, School of Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
121
|
Sandin JN, Aryal SP, Wilkop T, Richards CI, Grady ME. Near Simultaneous Laser Scanning Confocal and Atomic Force Microscopy (Conpokal) on Live Cells. J Vis Exp 2020:10.3791/61433. [PMID: 32865532 PMCID: PMC7680637 DOI: 10.3791/61433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Techniques available for micro- and nano-scale mechanical characterization have exploded in the last few decades. From further development of the scanning and transmission electron microscope, to the invention of atomic force microscopy, and advances in fluorescent imaging, there have been substantial gains in technologies that enable the study of small materials. Conpokal is a portmanteau that combines confocal microscopy with atomic force microscopy (AFM), where a probe "pokes" the surface. Although each technique is extremely effective for the qualitative and/or quantitative image collection on their own, Conpokal provides the capability to test with blended fluorescence imaging and mechanical characterization. Designed for near simultaneous confocal imaging and atomic force probing, Conpokal facilitates experimentation on live microbiological samples. The added insight from paired instrumentation provides co-localization of measured mechanical properties (e.g., elastic modulus, adhesion, surface roughness) by AFM with subcellular components or activity observable through confocal microscopy. This work provides a step by step protocol for the operation of laser scanning confocal and atomic force microscopy, simultaneously, to achieve same cell, same region, confocal imaging, and mechanical characterization.
Collapse
Affiliation(s)
- Joree N Sandin
- Department of Mechanical Engineering, University of Kentucky
| | | | - Thomas Wilkop
- Department of Physiology, University of Kentucky; UK Light Microscopy Core, University of Kentucky
| | - Christopher I Richards
- Department of Chemistry, University of Kentucky; UK Light Microscopy Core, University of Kentucky
| | - Martha E Grady
- Department of Mechanical Engineering, University of Kentucky;
| |
Collapse
|
122
|
Chtcheglova LA, Ohlmann A, Boytsov D, Hinterdorfer P, Priglinger SG, Priglinger CS. Nanoscopic Approach to Study the Early Stages of Epithelial to Mesenchymal Transition (EMT) of Human Retinal Pigment Epithelial (RPE) Cells In Vitro. Life (Basel) 2020; 10:E128. [PMID: 32751632 PMCID: PMC7460373 DOI: 10.3390/life10080128] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 12/23/2022] Open
Abstract
The maintenance of visual function is supported by the proper functioning of the retinal pigment epithelium (RPE), representing a mosaic of polarized cuboidal postmitotic cells. Damage factors such as inflammation, aging, or injury can initiate the migration and proliferation of RPE cells, whereas they undergo a pseudo-metastatic transformation or an epithelial to mesenchymal transition (EMT) from cuboidal epithelioid into fibroblast-like or macrophage-like cells. This process is recognized as a key feature in several severe ocular pathologies, and is mimicked by placing RPE cells in culture, which provides a reasonable and well-characterized in vitro model for a type 2 EMT. The most obvious characteristic of EMT is the cell phenotype switching, accompanied by the cytoskeletal reorganization with changes in size, shape, and geometry. Atomic force microscopy (AFM) has the salient ability to label-free explore these characteristics. Based on our AFM results supported by the genetic analysis of specific RPE differentiation markers, we elucidate a scheme for gradual transformation from the cobblestone to fibroblast-like phenotype. Structural changes in the actin cytoskeletal reorganization at the early stages of EMT lead to the development of characteristic geodomes, a finding that may reflect an increased propensity of RPE cells to undergo further EMT and thus become of diagnostic significance.
Collapse
Affiliation(s)
- Lilia A. Chtcheglova
- Institute of Biophysics, Johannes Kepler University (JKU) Linz, Gruberstrasse 40, 4020 Linz, Austria; (D.B.); (P.H.)
| | - Andreas Ohlmann
- Department of Ophthalmology, Munich University Hospital, Ludwig-Maximilians-University (LMU) Munich, Mathildenstrasse 8, 80336 Munich, Germany; (A.O.); (S.G.P.); (C.S.P.)
| | - Danila Boytsov
- Institute of Biophysics, Johannes Kepler University (JKU) Linz, Gruberstrasse 40, 4020 Linz, Austria; (D.B.); (P.H.)
| | - Peter Hinterdorfer
- Institute of Biophysics, Johannes Kepler University (JKU) Linz, Gruberstrasse 40, 4020 Linz, Austria; (D.B.); (P.H.)
| | - Siegfried G. Priglinger
- Department of Ophthalmology, Munich University Hospital, Ludwig-Maximilians-University (LMU) Munich, Mathildenstrasse 8, 80336 Munich, Germany; (A.O.); (S.G.P.); (C.S.P.)
| | - Claudia S. Priglinger
- Department of Ophthalmology, Munich University Hospital, Ludwig-Maximilians-University (LMU) Munich, Mathildenstrasse 8, 80336 Munich, Germany; (A.O.); (S.G.P.); (C.S.P.)
| |
Collapse
|
123
|
Zhou G, Zhang B, Wei L, Zhang H, Galluzzi M, Li J. Spatially Resolved Correlation between Stiffness Increase and Actin Aggregation around Nanofibers Internalized in Living Macrophages. MATERIALS 2020; 13:ma13143235. [PMID: 32708102 PMCID: PMC7412258 DOI: 10.3390/ma13143235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 12/21/2022]
Abstract
Plasticity and functional diversity of macrophages play an important role in resisting pathogens invasion, tumor progression and tissue repair. At present, nanodrug formulations are becoming increasingly important to induce and control the functional diversity of macrophages. In this framework, the internalization process of nanodrugs is co-regulated by a complex interplay of biochemistry, cell physiology and cell mechanics. From a biophysical perspective, little is known about cellular mechanics’ modulation induced by the nanodrug carrier’s internalization. In this study, we used the polylactic-co-glycolic acid (PLGA)–polyethylene glycol (PEG) nanofibers as a model drug carrier, and we investigated their influence on macrophage mechanics. Interestingly, the nanofibers internalized in macrophages induced a local increase of stiffness detected by atomic force microscopy (AFM) nanomechanical investigation. Confocal laser scanning microscopy revealed a thickening of actin filaments around nanofibers during the internalization process. Following geometry and mechanical properties by AFM, indentation experiments are virtualized in a finite element model simulation. It turned out that it is necessary to include an additional actin wrapping layer around nanofiber in order to achieve similar reaction force of AFM experiments, consistent with confocal observation. The quantitative investigation of actin reconfiguration around internalized nanofibers can be exploited to develop novel strategies for drug delivery.
Collapse
Affiliation(s)
- Guoqiao Zhou
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bokai Zhang
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
| | - Liyu Wei
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
| | - Han Zhang
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
- State Key Laboratory of Traction Power, Southwest Jiaotong University, Chengdu 610000, China
| | - Massimiliano Galluzzi
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
- Correspondence: (M.G.); (J.L.)
| | - Jiangyu Li
- Shenzhen Key Laboratory of Nanobiomechanics, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (G.Z.); (B.Z.); (L.W.); (H.Z.)
- Correspondence: (M.G.); (J.L.)
| |
Collapse
|
124
|
Khan AU, Qu R, Fan T, Ouyang J, Dai J. A glance on the role of actin in osteogenic and adipogenic differentiation of mesenchymal stem cells. Stem Cell Res Ther 2020; 11:283. [PMID: 32678016 PMCID: PMC7364498 DOI: 10.1186/s13287-020-01789-2] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/13/2020] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have the capacity to differentiate into multiple lineages including osteogenic and adipogenic lineages. An increasing number of studies have indicated that lineage commitment by MSCs is influenced by actin remodeling. Moreover, actin has roles in determining cell shape, nuclear shape, cell spreading, and cell stiffness, which eventually affect cell differentiation. Osteogenic differentiation is promoted in MSCs that exhibit a large spreading area, increased matrix stiffness, higher levels of actin polymerization, and higher density of stress fibers, whereas adipogenic differentiation is prevalent in MSCs with disrupted actin networks. In addition, the mechanical properties of F-actin empower cells to sense and transduce mechanical stimuli, which are also reported to influence differentiation. Various biomaterials, mechanical, and chemical interventions along with pathogen-induced actin alteration in the form of polymerization and depolymerization in MSC differentiation were studied recently. This review will cover the role of actin and its modifications through the use of different methods in inducing osteogenic and adipogenic differentiation.
Collapse
Affiliation(s)
- Asmat Ullah Khan
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Rongmei Qu
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Tingyu Fan
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jun Ouyang
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Jingxing Dai
- Guangdong Provincial Key Laboratory of Medical Biomechanics, Department of Anatomy, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| |
Collapse
|
125
|
Huang W, Tan M, Wang Y, Liu L, Pan Y, Li J, Ouyang M, Long C, Qu X, Liu H, Liu C, Wang J, Deng L, Xiang Y, Qin X. Increased intracellular Cl - concentration improves airway epithelial migration by activating the RhoA/ROCK Pathway. Theranostics 2020; 10:8528-8540. [PMID: 32754261 PMCID: PMC7392015 DOI: 10.7150/thno.46002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022] Open
Abstract
In the airway, Cl- is the most abundant anion and is critically involved in transepithelial transport. The correlation of the abnormal expression and activation of chloride channels (CLCs), such as cystic fibrosis transmembrane conductance regulators (CFTRs), anoctamin-1, and CLC-2, with cell migration capability suggests a relationship between defective Cl- transport and epithelial wound repair. However, whether a correlation exists between intracellular Cl- and airway wound repair capability has not been explored thus far, and the underlying mechanisms involved in this relationship are not fully defined. Methods: In this work, the alteration of intracellular chloride concentration ([Cl-]i) was measured by using a chloride-sensitive fluorescent probe (N-[ethoxycarbonylmethyl]-6-methoxyquinolium bromide). Results: We found that clamping with high [Cl-]i and 1 h of treatment with the CLC inhibitor CFTR blocker CFTRinh-172 and chloride intracellular channel inhibitor IAA94 increased intracellular Cl- concentration ([Cl-]i) in airway epithelial cells. This effect improved epithelial cell migration. In addition, increased [Cl-]i in cells promoted F-actin reorganization, decreased cell stiffness, and improved RhoA activation and LIMK1/2 phosphorylation. Treatment with the ROCK inhibitor of Y-27632 and ROCK1 siRNA significantly attenuated the effects of increased [Cl-]i on LIMK1/2 activation and cell migration. In addition, intracellular Ca2+ concentration was unaffected by [Cl-]i clamping buffers and CFTRinh-172 and IAA94. Conclusion: Taken together, these results suggested that Cl- accumulation in airway epithelial cells could activate the RhoA/ROCK/LIMK cascade to induce F-actin reorganization, down-regulate cell stiffness, and improve epithelial migration.
Collapse
Affiliation(s)
- Wenjie Huang
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
- Affiliated Liutie Central Hospital of Guangxi medical university, Liuzhou, Guangxi 545007, China
| | - Meiling Tan
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Yue Wang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, Jiangsu 213164, China
- School of Nursing, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Lei Liu
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, Jiangsu 213164, China
| | - Yan Pan
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, Jiangsu 213164, China
| | - Jingjing Li
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, Jiangsu 213164, China
| | - Mingxing Ouyang
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, Jiangsu 213164, China
| | - Chunjiao Long
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xiangping Qu
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Huijun Liu
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Chi Liu
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Jia Wang
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Linhong Deng
- Changzhou Key Laboratory of Respiratory Medical Engineering, Institute of Biomedical Engineering and Health Sciences, Changzhou, Jiangsu 213164, China
| | - Yang Xiang
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| | - Xiaoqun Qin
- School of Basic Medicine, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
126
|
Desbiolles BXE, Hannebelle MTM, de Coulon E, Bertsch A, Rohr S, Fantner GE, Renaud P. Volcano-Shaped Scanning Probe Microscopy Probe for Combined Force-Electrogram Recordings from Excitable Cells. NANO LETTERS 2020; 20:4520-4529. [PMID: 32426984 PMCID: PMC7291358 DOI: 10.1021/acs.nanolett.0c01319] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/19/2020] [Indexed: 05/30/2023]
Abstract
Atomic force microscopy based approaches have led to remarkable advances in the field of mechanobiology. However, linking the mechanical cues to biological responses requires complementary techniques capable of recording these physiological characteristics. In this study, we present an instrument for combined optical, force, and electrical measurements based on a novel type of scanning probe microscopy cantilever composed of a protruding volcano-shaped nanopatterned microelectrode (nanovolcano probe) at the tip of a suspended microcantilever. This probe enables simultaneous force and electrical recordings from single cells. Successful impedance measurements on mechanically stimulated neonatal rat cardiomyocytes in situ were achieved using these nanovolcano probes. Furthermore, proof of concept experiments demonstrated that extracellular field potentials (electrogram) together with contraction displacement curves could simultaneously be recorded. These features render the nanovolcano probe especially suited for mechanobiological studies aiming at linking mechanical stimuli to electrophysiological responses of single cells.
Collapse
Affiliation(s)
- B. X. E. Desbiolles
- Laboratory
of Microsystems LMIS4, Ecole Polytechnique
Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - M. T. M Hannebelle
- Laboratory
of Bio- and Nano- Instrumentation, Ecole
Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - E. de Coulon
- Laboratory
of Cellular Optics II, Department of Physiology, University of Bern, Bern 3012, Switzerland
| | - A. Bertsch
- Laboratory
of Microsystems LMIS4, Ecole Polytechnique
Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - S. Rohr
- Laboratory
of Cellular Optics II, Department of Physiology, University of Bern, Bern 3012, Switzerland
| | - G. E. Fantner
- Laboratory
of Bio- and Nano- Instrumentation, Ecole
Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - P. Renaud
- Laboratory
of Microsystems LMIS4, Ecole Polytechnique
Fédérale de Lausanne, Lausanne 1015, Switzerland
| |
Collapse
|
127
|
Hohmann T, Hohmann U, Kolbe MR, Dahlmann M, Kobelt D, Stein U, Dehghani F. MACC1 driven alterations in cellular biomechanics facilitate cell motility in glioblastoma. Cell Commun Signal 2020; 18:85. [PMID: 32503676 PMCID: PMC7275321 DOI: 10.1186/s12964-020-00566-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 03/23/2020] [Indexed: 12/31/2022] Open
Abstract
Background Metastasis-associated in colon cancer 1 (MACC1) is an established marker for metastasis and tumor cell migration in a multitude of tumor entities, including glioblastoma (GBM). Nevertheless, the mechanism underlying the increased migratory capacity in GBM is not comprehensively explored. Methods We performed live cell and atomic force microscopy measurements to assess cell migration and mechanical properties of MACC1 overexpressing GBM cells. We quantified MACC1 dependent dynamics of 3D aggregate formation. For mechanistic studies we measured the expression of key adhesion molecules using qRT-PCR, and MACC1 dependent changes in short term adhesion to fibronectin and laminin. We then determined changes in sub-cellular distribution of integrins and actin in dependence of MACC1, but also in microtubule and intermediate filament organization. Results MACC1 increased the migratory speed and elastic modulus of GBM cells, but decreased cell-cell adhesion and inhibited the formation of 3D aggregates. These effects were not associated with altered mRNA expression of several key adhesion molecules or altered short-term affinity to laminin and fibronectin. MACC1 did neither change the organization of the microtubule nor intermediate filament cytoskeleton, but resulted in increased amounts of protrusive actin on laminin. Conclusion MACC1 overexpression increases elastic modulus and migration and reduces adhesion of GBM cells thereby impeding 3D aggregate formation. The underlying molecular mechanism is independent on the organization of microtubules, intermediate filaments and several key adhesion molecules, but depends on adhesion to laminin. Thus, targeting re-organization of the cytoskeleton and cell motility via MACC1 may offer a treatment option to impede GBM spreading. Video Abstract
Collapse
Affiliation(s)
- Tim Hohmann
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108, Halle, Saale, Germany
| | - Urszula Hohmann
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108, Halle, Saale, Germany
| | - Marc R Kolbe
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108, Halle, Saale, Germany
| | - Mathias Dahlmann
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Dennis Kobelt
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany.,German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany. .,German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| | - Faramarz Dehghani
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108, Halle, Saale, Germany.
| |
Collapse
|
128
|
Del Favero G, Kraegeloh A. Integrating Biophysics in Toxicology. Cells 2020; 9:E1282. [PMID: 32455794 PMCID: PMC7290780 DOI: 10.3390/cells9051282] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/10/2020] [Accepted: 05/15/2020] [Indexed: 12/20/2022] Open
Abstract
Integration of biophysical stimulation in test systems is established in diverse branches of biomedical sciences including toxicology. This is largely motivated by the need to create novel experimental setups capable of reproducing more closely in vivo physiological conditions. Indeed, we face the need to increase predictive power and experimental output, albeit reducing the use of animals in toxicity testing. In vivo, mechanical stimulation is essential for cellular homeostasis. In vitro, diverse strategies can be used to model this crucial component. The compliance of the extracellular matrix can be tuned by modifying the stiffness or through the deformation of substrates hosting the cells via static or dynamic strain. Moreover, cells can be cultivated under shear stress deriving from the movement of the extracellular fluids. In turn, introduction of physical cues in the cell culture environment modulates differentiation, functional properties, and metabolic competence, thus influencing cellular capability to cope with toxic insults. This review summarizes the state of the art of integration of biophysical stimuli in model systems for toxicity testing, discusses future challenges, and provides perspectives for the further advancement of in vitro cytotoxicity studies.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Währinger Straße 38-40, 1090 Vienna, Austria
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna Währinger Straße 38-40, 1090 Vienna, Austria
| | - Annette Kraegeloh
- INM—Leibniz-Institut für Neue Materialien GmbH, Campus D2 2, 66123 Saarbrücken, Germany;
| |
Collapse
|
129
|
Nguyen TL, Polanco ER, Patananan AN, Zangle TA, Teitell MA. Cell viscoelasticity is linked to fluctuations in cell biomass distributions. Sci Rep 2020; 10:7403. [PMID: 32366921 PMCID: PMC7198624 DOI: 10.1038/s41598-020-64259-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/14/2020] [Indexed: 12/20/2022] Open
Abstract
The viscoelastic properties of mammalian cells can vary with biological state, such as during the epithelial-to-mesenchymal (EMT) transition in cancer, and therefore may serve as a useful physical biomarker. To characterize stiffness, conventional techniques use cell contact or invasive probes and as a result are low throughput, labor intensive, and limited by probe placement. Here, we show that measurements of biomass fluctuations in cells using quantitative phase imaging (QPI) provides a probe-free, contact-free method for quantifying changes in cell viscoelasticity. In particular, QPI measurements reveal a characteristic underdamped response of changes in cell biomass distributions versus time. The effective stiffness and viscosity values extracted from these oscillations in cell biomass distributions correlate with effective cell stiffness and viscosity measured by atomic force microscopy (AFM). This result is consistent for multiple cell lines with varying degrees of cytoskeleton disruption and during the EMT. Overall, our study demonstrates that QPI can reproducibly quantify cell viscoelasticity.
Collapse
Affiliation(s)
- Thang L Nguyen
- Department of Bioengineering, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Edward R Polanco
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Alexander N Patananan
- Deparment of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Thomas A Zangle
- Department of Chemical Engineering, University of Utah, Salt Lake City, UT, 84112, USA.
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Michael A Teitell
- Department of Bioengineering, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
- Deparment of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
- Broad Center for Regenerative Medicine and Stem Cell Research, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
- California NanoSystems Institute, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Pediatrics, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
130
|
Zhang J, Alisafaei F, Nikolić M, Nou XA, Kim H, Shenoy VB, Scarcelli G. Nuclear Mechanics within Intact Cells Is Regulated by Cytoskeletal Network and Internal Nanostructures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907688. [PMID: 32243075 PMCID: PMC7799396 DOI: 10.1002/smll.201907688] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 03/05/2020] [Accepted: 03/05/2020] [Indexed: 05/11/2023]
Abstract
The mechanical properties of the cellular nucleus are extensively studied as they play a critical role in important processes, such as cell migration, gene transcription, and stem cell differentiation. While the mechanical properties of the isolated nucleus have been tested, there is a lack of measurements about the mechanical behavior of the nucleus within intact cells and specifically about the interplay of internal nuclear components with the intracellular microenvironment, because current testing methods are based on contact and only allow studying the nucleus after isolation from a cell or disruption of cytoskeleton. Here, all-optical Brillouin microscopy and 3D chemomechanical modeling are used to investigate the regulation of nuclear mechanics in physiological conditions. It is observed that the nuclear modulus can be modulated by epigenetic regulation targeting internal nuclear nanostructures such as lamin A/C and chromatin. It is also found that nuclear modulus is strongly regulated by cytoskeletal behavior through a robust mechanism conserved in different culturing conditions. Given the active role of cytoskeletal modulation in nearly all cell functions, this work will enable to reveal highly relevant mechanisms of nuclear mechanical regulations in physiological and pathological conditions.
Collapse
Affiliation(s)
- Jitao Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Farid Alisafaei
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, PA, 19104, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, PA, 19104, USA
| | - Miloš Nikolić
- Maryland Biophysics Program, University of Maryland, College Park, MD 20742, USA
| | - Xuefei A. Nou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Hanyoup Kim
- Canon U.S. Life Sciences, Inc., 9800 Medical Center Drive, Suite C-120, Rockville, MD 20850, USA
| | - Vivek B. Shenoy
- Department of Materials Science and Engineering, School of Engineering and Applied Science, University of Pennsylvania, PA, 19104, USA
- Center for Engineering Mechanobiology, University of Pennsylvania, PA, 19104, USA
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Maryland Biophysics Program, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
131
|
Graybill PM, Davalos RV. Cytoskeletal Disruption after Electroporation and Its Significance to Pulsed Electric Field Therapies. Cancers (Basel) 2020; 12:E1132. [PMID: 32366043 PMCID: PMC7281591 DOI: 10.3390/cancers12051132] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Pulsed electric fields (PEFs) have become clinically important through the success of Irreversible Electroporation (IRE), Electrochemotherapy (ECT), and nanosecond PEFs (nsPEFs) for the treatment of tumors. PEFs increase the permeability of cell membranes, a phenomenon known as electroporation. In addition to well-known membrane effects, PEFs can cause profound cytoskeletal disruption. In this review, we summarize the current understanding of cytoskeletal disruption after PEFs. Compiling available studies, we describe PEF-induced cytoskeletal disruption and possible mechanisms of disruption. Additionally, we consider how cytoskeletal alterations contribute to cell-cell and cell-substrate disruption. We conclude with a discussion of cytoskeletal disruption-induced anti-vascular effects of PEFs and consider how a better understanding of cytoskeletal disruption after PEFs may lead to more effective therapies.
Collapse
Affiliation(s)
- Philip M. Graybill
- BEMS Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA;
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Rafael V. Davalos
- BEMS Lab, Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA 24061, USA;
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA 24061, USA
- Virginia Tech–Wake Forest University, School of Biomedical Engineering and Sciences, Blacksburg, VA 24061, USA
| |
Collapse
|
132
|
Garcia PD, Guerrero CR, Garcia R. Nanorheology of living cells measured by AFM-based force-distance curves. NANOSCALE 2020; 12:9133-9143. [PMID: 32293616 DOI: 10.1039/c9nr10316c] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Mechanobiology aims to establish functional relationships between the mechanical state of a living a cell and its physiology. The acquisition of force-distance curves with an AFM is by far the dominant method to characterize the nanomechanical properties of living cells. However, theoretical simulations have shown that the contact mechanics models used to determine the Young's modulus from a force-distance curve could be off by a factor 5 from its expected value. The semi-quantitative character arises from the lack of a theory that integrates the AFM data, a realistic viscoelastic model of a cell and its finite-thickness. Here, we develop a method to determine the mechanical response of a cell from a force-distance curve. The method incorporates bottom-effect corrections, a power-law rheology model and the deformation history of the cell. It transforms the experimental data into viscoelastic parameters of the cell as a function of the indentation frequency. The quantitative agreement obtained between the experiments performed on living fibroblast cells and the analytical theory supports the use of force-distance curves to measure the nanorheological properties of cells.
Collapse
Affiliation(s)
- Pablo D Garcia
- Instituto de Ciencia de Materiales de Madrid, CSIC, c/ Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain.
| | | | | |
Collapse
|
133
|
Yen MH, Chen YH, Liu YS, Lee OKS. Alteration of Young's modulus in mesenchymal stromal cells during osteogenesis measured by atomic force microscopy. Biochem Biophys Res Commun 2020; 526:827-832. [PMID: 32273088 DOI: 10.1016/j.bbrc.2020.03.146] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 03/25/2020] [Indexed: 12/22/2022]
Abstract
Mechanical properties of biological tissues are increasingly recognized as an important parameter for the indication of disease states as well as tissue homeostasis and regeneration. Multipotent mesenchymal stromal/stem cells (MSCs), which play important roles in bone formation and remodeling, are potential cell sources for regenerative medicine. However, the cellular mechanical properties of differentiating MSCs corresponding to the substrate stiffness has not been sufficiently studied. In this study, we used Atomic Force Microscopy (AFM) to measure changes of stiffness of human MSCs cultured in rigid Petri dish and on polyacrylamide (PA) substrates during osteogenic differentiation. The results showed that the Young's modulus of MSC cytoplasmic outer region increased over time during osteogenesis. There is a strong linear correlation between the osteogenic induction time and the Young's modulus of the cells cultured in rigid Petri dishes in the first 15 days after the induction; the Young's modulus approaches to a plateau after day 15. On the other hand, the Young's moduli of MSCs cultured on PA gels with stiffness of 7 kPa and 42 kPa also increase over time during osteogenic differentiation, but the inclination of such increase is much smaller than that of MSCs differentiating in rigid dishes. Herein, we established a protocol of AFM measurement to evaluate the maturation of stem cell osteogenic differentiation at the single cell level and could encourage further AFM applications in tissue engineering related to mechanobiology.
Collapse
Affiliation(s)
- Meng-Hua Yen
- Institute of Clinical Medicine, and Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Electronic Engineering, National Chin-Yi University of Technology, Taichung, Taiwan.
| | - Yu-Han Chen
- Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Shiuan Liu
- Institute of Clinical Medicine, and Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Physiology and Pharmacology, Chang Gung University College of Medicine, and Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| | - Oscar Kuang-Sheng Lee
- Institute of Clinical Medicine, and Stem Cell Research Center, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.
| |
Collapse
|
134
|
Kwon S, Kim KS. Qualitative analysis of contribution of intracellular skeletal changes to cellular elasticity. Cell Mol Life Sci 2020; 77:1345-1355. [PMID: 31605149 PMCID: PMC11105102 DOI: 10.1007/s00018-019-03328-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 01/07/2023]
Abstract
Cells are dynamic structures that continually generate and sustain mechanical forces within their environments. Cells respond to mechanical forces by changing their shape, moving, and differentiating. These reactions are caused by intracellular skeletal changes, which induce changes in cellular mechanical properties such as stiffness, elasticity, viscoelasticity, and adhesiveness. Interdisciplinary research combining molecular biology with physics and mechanical engineering has been conducted to characterize cellular mechanical properties and understand the fundamental mechanisms of mechanotransduction. In this review, we focus on the role of cytoskeletal proteins in cellular mechanics. The specific role of each cytoskeletal protein, including actin, intermediate filaments, and microtubules, on cellular elasticity is summarized along with the effects of interactions between the fibers.
Collapse
Affiliation(s)
- Sangwoo Kwon
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Kyung Sook Kim
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
135
|
Rosowski KA, Sai T, Vidal-Henriquez E, Zwicker D, Style RW, Dufresne ER. Elastic ripening and inhibition of liquid-liquid phase separation. NATURE PHYSICS 2020; 16:422-425. [PMID: 32273899 PMCID: PMC7145441 DOI: 10.1038/s41567-019-0767-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/02/2019] [Indexed: 05/07/2023]
Abstract
Phase separation is a central concept of materials physics [1-3] and has recently emerged as an important route to compartmentalization within living cells [4-6]. Biological phase separation features activity [7], complex compositions [8], and elasticity [9], which reveal important gaps in our understanding of this universal physical phenomenon. Here, we explore the impact of elasticity on phase separation in synthetic polymer networks. We show that compressive stresses in a polymer network can suppress phase separation of the solvent that swells it, stabilizing mixtures well beyond the liquid-liquid phase separation boundary. Network stresses also drive a new form of ripening, driven by transport of solute down stiffness gradients. This elastic ripening can be much faster than conventional surface tension driven Ostwald ripening.
Collapse
Affiliation(s)
| | - Tianqi Sai
- Department of Materials, ETH Zürich, 8093 Zürich, Switzerland
| | | | - David Zwicker
- Max Planck Institute for Dynamics and Self-Organization, 37077, Göttingen, Germany
| | - Robert W. Style
- Department of Materials, ETH Zürich, 8093 Zürich, Switzerland
| | | |
Collapse
|
136
|
Bui VC, Nguyen TH. Direct monitoring of drug-induced mechanical response of individual cells by atomic force microscopy. J Mol Recognit 2020; 33:e2847. [PMID: 32212218 DOI: 10.1002/jmr.2847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 03/08/2020] [Accepted: 03/10/2020] [Indexed: 01/12/2023]
Abstract
Mechanical characteristics of individual cells play a vital role in many biological processes and are considered as indicators of the cells' states. Disturbances including methyl-β-cyclodextrin (MβCD) and cytochalasin D (cytoD) are known to significantly affect the state of cells, but little is known about the real-time response of single cells to these drugs in their physiological condition. Here, nanoindentation-based atomic force microscopy (AFM) was used to measure the elasticity of human embryonic kidney cells in the presence and absence of these pharmaceuticals. The results showed that depletion of cholesterol in the plasma membrane with MβCD resulted in cell stiffening whereas depolymerization of the actin cytoskeleton by cytoD resulted in cell softening. Using AFM for real-time measurements, we observed that cells mechanically responded right after these drugs were added. In more detail, the cell´s elasticity suddenly increased with increasing instability upon cholesterol extraction while it is rapidly decreased without changing cellular stability upon depolymerizing actin cytoskeleton. These results demonstrated that actin cytoskeleton and cholesterol contributed differently to the cell mechanical characteristics.
Collapse
Affiliation(s)
- Van-Chien Bui
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany.,ZIK HIKE, University of Greifswald, Greifswald, Germany
| | - Thi-Huong Nguyen
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany.,Institute for Bioprocessing and Analytical Measurement Techniques, Heilbad Heiligenstadt, Germany
| |
Collapse
|
137
|
Lv JQ, Chen PC, Góźdź WT, Li B. Mechanical adaptions of collective cells nearby free tissue boundaries. J Biomech 2020; 104:109763. [PMID: 32224050 DOI: 10.1016/j.jbiomech.2020.109763] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 11/19/2022]
Abstract
Mechanical adaptions of cells, including stiffness variation, cytoskeleton remodeling, motion coordination, and shape changing, are essential for tissue morphogenesis, wound healing, and malignant progression. In this paper, we take confluent monolayers of Madin-Darby canine kidney (MDCK) and mouse myoblast (C2C12) cells as model systems to probe how cells collectively adapt their mechanical features in response to a free tissue boundary. We show that the free boundary not only can trigger unjamming transition but also induces cell fluidization nearby the boundary. The Young's moduli of cells near the boundary are found to be much lower than those of interior cells. We demonstrate that the stiffness of cells in monolayers with a free tissue boundary exhibits negative dependence on the projected cell area, in contrast to previous studies where cells were found to stiffen as cellular area increases in a confluent MDCK monolayer without boundary. In addition, the free tissue boundary may activate cell remodeling, rendering volume enlargement and cell-specified cytoskeleton organization. Our study emphasizes the important role of geometrical boundary in regulating biomechanical properties of cell aggregates.
Collapse
Affiliation(s)
- Jian-Qing Lv
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Peng-Cheng Chen
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Wojciech T Góźdź
- Institute of Physical Chemistry, Polish Academy of Sciences, Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - Bo Li
- Institute of Biomechanics and Medical Engineering, Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
138
|
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: 29] [Impact Index Per Article: 5.8] [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
|
139
|
Effect of F-actin and Microtubules on Cellular Mechanical Behavior Studied Using Atomic Force Microscope and an Image Recognition-Based Cytoskeleton Quantification Approach. Int J Mol Sci 2020; 21:ijms21020392. [PMID: 31936268 PMCID: PMC7014474 DOI: 10.3390/ijms21020392] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 11/16/2022] Open
Abstract
Cytoskeleton morphology plays a key role in regulating cell mechanics. Particularly, cellular mechanical properties are directly regulated by the highly cross-linked and dynamic cytoskeletal structure of F-actin and microtubules presented in the cytoplasm. Although great efforts have been devoted to investigating the qualitative relation between the cellular cytoskeleton state and cell mechanical properties, comprehensive quantification results of how the states of F-actin and microtubules affect mechanical behavior are still lacking. In this study, the effect of both F-actin and microtubules morphology on cellular mechanical properties was quantified using atomic force microscope indentation experiments together with the proposed image recognition-based cytoskeleton quantification approach. Young’s modulus and diffusion coefficient of NIH/3T3 cells with different cytoskeleton states were quantified at different length scales. It was found that the living NIH/3T3 cells sense and adapt to the F-actin and microtubules states: both the cellular elasticity and poroelasticity are closely correlated to the depolymerization degree of F-actin and microtubules at all measured indentation depths. Moreover, the significance of the quantitative effects of F-actin and microtubules in affecting cellular mechanical behavior is depth-dependent.
Collapse
|
140
|
Efremov YM, Okajima T, Raman A. Measuring viscoelasticity of soft biological samples using atomic force microscopy. SOFT MATTER 2020; 16:64-81. [PMID: 31720656 DOI: 10.1039/c9sm01020c] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mechanical properties play important roles at different scales in biology. At the level of a single cell, the mechanical properties mediate mechanosensing and mechanotransduction, while at the tissue and organ levels, changes in mechanical properties are closely connected to disease and physiological processes. Over the past three decades, atomic force microscopy (AFM) has become one of the most widely used tools in the mechanical characterization of soft samples, ranging from molecules, cell organoids and cells to whole tissue. AFM methods can be used to quantify both elastic and viscoelastic properties, and significant recent developments in the latter have been enabled by the introduction of new techniques and models for data analysis. Here, we review AFM techniques developed in recent years for examining the viscoelastic properties of cells and soft gels, describe the main steps in typical data acquisition and analysis protocols, and discuss relevant viscoelastic models and how these have been used to characterize the specific features of cellular and other biological samples. We also discuss recent trends and potential directions for this field.
Collapse
Affiliation(s)
- Yuri M Efremov
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA. and Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana, USA and Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Takaharu Okajima
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
| | - Arvind Raman
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana, USA. and Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
141
|
Wala J, Das S. Mapping of biomechanical properties of cell lines on altered matrix stiffness using atomic force microscopy. Biomech Model Mechanobiol 2020; 19:1523-1536. [DOI: 10.1007/s10237-019-01285-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 12/28/2019] [Indexed: 01/07/2023]
|
142
|
Wu Y, Cheng T, Chen Q, Gao B, Stewart AG, Lee PVS. On-chip surface acoustic wave and micropipette aspiration techniques to assess cell elastic properties. BIOMICROFLUIDICS 2020; 14:014114. [PMID: 32095200 PMCID: PMC7028434 DOI: 10.1063/1.5138662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/07/2020] [Indexed: 05/26/2023]
Abstract
The cytoskeletal mechanics and cell mechanical properties play an important role in cellular behaviors. In this study, in order to provide comprehensive insights into the relationship between different cytoskeletal components and cellular elastic moduli, we built a phase-modulated surface acoustic wave microfluidic device to measure cellular compressibility and a microfluidic micropipette-aspiration device to measure cellular Young's modulus. The microfluidic devices were validated based on experimental data and computational simulations. The contributions of structural cytoskeletal actin filament and microtubule to cellular compressibility and Young's modulus were examined in MCF-7 cells. The compressibility of MCF-7 cells was increased after microtubule disruption, whereas actin disruption had no effect. In contrast, Young's modulus of MCF-7 cells was reduced after actin disruption but unaffected by microtubule disruption. The actin filaments and microtubules were stained to confirm the structural alteration in cytoskeleton. Our findings suggest the dissimilarity in the structural roles of actin filaments and microtubules in terms of cellular compressibility and Young's modulus. Based on the differences in location and structure, actin filaments mainly contribute to tensile Young's modulus and microtubules mainly contribute to compressibility. In addition, different responses to cytoskeletal alterations between acoustophoresis and micropipette aspiration demonstrated that micropipette aspiration was better at detecting the change from actin cortex, while the response to acoustophoresis was governed by microtubule networks.
Collapse
Affiliation(s)
- Yanqi Wu
- Department of Biomedical Engineering, University of Melbourne, Melbourne, VIC 3010, Australia
| | | | | | | | | | - Peter V. S. Lee
- Department of Biomedical Engineering, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
143
|
Saidova AA, Vorobjev IA. Lineage Commitment, Signaling Pathways, and the Cytoskeleton Systems in Mesenchymal Stem Cells. TISSUE ENGINEERING PART B-REVIEWS 2019; 26:13-25. [PMID: 31663422 DOI: 10.1089/ten.teb.2019.0250] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) from adult tissues are promising candidates for personalized cell therapy and tissue engineering. Significant progress was achieved in our understanding of the regulation of MSCs proliferation and differentiation by different cues during the past years. Proliferation and differentiation of MSCs are sensitive to the extracellular matrix (ECM) properties, physical cues, and chemical signaling. Sheath stress, matrix stiffness, surface adhesiveness, and micro- and nanotopography define cell shape and dictate lineage commitment of MSCs even in the absence of specific chemical signals. We discuss mechanotransduction as the major route from ECM through the cytoskeleton toward signaling pathways and gene expression. All components of the cytoskeleton from primary cilium and focal adhesions (FAs) to actin, microtubules (MTs), and intermediate filaments (IFs) are involved in the mechanotransduction. Differentiation of MSCs is regulated via the complex network of interrelated signaling pathways, including RhoA/ROCK, Akt/Erk, and YAP/TAZ effectors of Hippo pathway. These pathways could be regulated both by chemical and mechanical stimuli. Attenuation of these pathways in MSCs results in specific changes in FAs and actin cytoskeleton. Besides, differentiation of MSCs affects MTs and IFs. Recent findings highlight the role of intranuclear actin in the regulation of transcription factors in response to mechanical environmental stimuli. Alterations of cytoskeletal components reflect the MSC senescence state and their migratory capacity. In this review, we discuss the relationships between the molecular interactions in signaling pathways and morphological response of cytoskeletal components and reveal the complex interrelations between cytoskeleton systems and signaling pathways during lineage commitment of MSCs. Impact Statement This review describes the complex network of relationships between mechanical and biochemical stimuli in mesenchymal stem cells (MSC) and their balance which defines the morphological changes of cell shape due to rearrangement of cytoskeletal systems during lineage commitment of MSCs.
Collapse
Affiliation(s)
- Aleena A Saidova
- Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia.,Center of Experimental Embryology and Reproductive Biotechnology, Moscow, Russia
| | - Ivan A Vorobjev
- Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia.,A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Department of Biology, School of Science and Humanities and National Laboratory Astana, Nazarbayev University, Nur-Sultan, Kazakhstan
| |
Collapse
|
144
|
Pires RH, Shree N, Manu E, Guzniczak E, Otto O. Cardiomyocyte mechanodynamics under conditions of actin remodelling. Philos Trans R Soc Lond B Biol Sci 2019; 374:20190081. [PMID: 31587648 PMCID: PMC6792454 DOI: 10.1098/rstb.2019.0081] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2019] [Indexed: 01/26/2023] Open
Abstract
The mechanical performance of cardiomyocytes (CMs) is an important indicator of their maturation state and of primary importance for the development of therapies based on cardiac stem cells. As the mechanical analysis of adherent cells at high-throughput remains challenging, we explore the applicability of real-time deformability cytometry (RT-DC) to probe cardiomyocytes in suspension. RT-DC is a microfluidic technology allowing for real-time mechanical analysis of thousands of cells with a throughput exceeding 1000 cells per second. For CMs derived from human-induced pluripotent stem cells, we determined a Young's modulus of 1.25 ± 0.08 kPa which is in close range to previous reports. Upon challenging the cytoskeleton with cytochalasin D (CytoD) to induce filamentous actin depolymerization, we distinguish three different regimes in cellular elasticity. Transitions are observed below 10 nM and above 103 nM and are characterized by a decrease in Young's modulus. These regimes can be linked to cytoskeletal and sarcomeric actin contributions by CM contractility measurements at varying CytoD concentrations, where we observe a significant reduction in pulse duration only above 103 nM while no change is found for compound exposure at lower concentrations. Comparing our results to mechanical cell measurements using atomic force microscopy, we demonstrate for the first time to our knowledge, the feasibility of using a microfluidic technique to measure mechanical properties of large samples of adherent cells while linking our results to the composition of the cytoskeletal network. This article is part of a discussion meeting issue 'Single cell ecology'.
Collapse
Affiliation(s)
- Ricardo H. Pires
- Zentrum für Innovationskompetenz: Humorale Immunreaktionen bei kardiovaskulären Erkrankungen, Universität Greifswald, Fleischmannstrasse 42, 17489 Greifswald, Germany
| | - Nithya Shree
- Zentrum für Innovationskompetenz: Humorale Immunreaktionen bei kardiovaskulären Erkrankungen, Universität Greifswald, Fleischmannstrasse 42, 17489 Greifswald, Germany
| | - Emmanuel Manu
- Zentrum für Innovationskompetenz: Humorale Immunreaktionen bei kardiovaskulären Erkrankungen, Universität Greifswald, Fleischmannstrasse 42, 17489 Greifswald, Germany
| | - Ewa Guzniczak
- Heriot-Watt University School of Engineering and Physical Science, Institute of Biological Chemistry, Biophysics and Bioengineering, Edinburgh Campus, Edinburgh EH14 4AS, UK
| | - Oliver Otto
- Zentrum für Innovationskompetenz: Humorale Immunreaktionen bei kardiovaskulären Erkrankungen, Universität Greifswald, Fleischmannstrasse 42, 17489 Greifswald, Germany
| |
Collapse
|
145
|
Yasunaga A, Murad Y, Li ITS. Quantifying molecular tension-classifications, interpretations and limitations of force sensors. Phys Biol 2019; 17:011001. [PMID: 31387091 DOI: 10.1088/1478-3975/ab38ff] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Molecular force sensors (MFSs) have grown to become an important tool to study the mechanobiology of cells and tissues. They provide a minimally invasive means to optically report mechanical interactions at the molecular level. One of the challenges in molecular force sensor studies is the interpretation of the fluorescence readout. In this review, we divide existing MFSs into three classes based on the force-sensing mechanism (reversibility) and the signal output (analog/digital). From single-molecule force spectroscopy (SMFS) perspectives, we provided a critical discussion on how the sensors respond to force and how the different sensor designs affect the interpretation of their fluorescence readout. Lastly, the review focuses on the limitations and attention one must pay in designing MFSs and biological experiments using them; in terms of their tunability, signal-to-noise ratio (SNR), and perturbation of the biological system under investigation.
Collapse
Affiliation(s)
- Adam Yasunaga
- These authors contributed equally to the manuscript (co-first author)
| | | | | |
Collapse
|
146
|
Weber A, Iturri J, Benitez R, Zemljic-Jokhadar S, Toca-Herrera JL. Microtubule disruption changes endothelial cell mechanics and adhesion. Sci Rep 2019; 9:14903. [PMID: 31624281 PMCID: PMC6797797 DOI: 10.1038/s41598-019-51024-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/24/2019] [Indexed: 12/27/2022] Open
Abstract
The interest in studying the mechanical and adhesive properties of cells has increased in recent years. The cytoskeleton is known to play a key role in cell mechanics. However, the role of the microtubules in shaping cell mechanics is not yet well understood. We have employed Atomic Force Microscopy (AFM) together with confocal fluorescence microscopy to determine the role of microtubules in cytomechanics of Human Umbilical Vein Endothelial Cells (HUVECs). Additionally, the time variation of the adhesion between tip and cell surface was studied. The disruption of microtubules by exposing the cells to two colchicine concentrations was monitored as a function of time. Already, after 30 min of incubation the cells stiffened, their relaxation times increased (lower fluidity) and the adhesion between tip and cell decreased. This was accompanied by cytoskeletal rearrangements, a reduction in cell area and changes in cell shape. Over the whole experimental time, different behavior for the two used concentrations was found while for the control the values remained stable. This study underlines the role of microtubules in shaping endothelial cell mechanics.
Collapse
Affiliation(s)
- Andreas Weber
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 11, A-1190, Vienna, Austria.
| | - Jagoba Iturri
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 11, A-1190, Vienna, Austria
| | - Rafael Benitez
- Dpto. Matemáticas para la Economía y la Empresa, Facultad de Economía, Universidad de Valencia, Avda. Tarongers s/n, 46022, Valencia, Spain
| | - Spela Zemljic-Jokhadar
- Department of Biophysics, Medicine Faculty, University of Ljubljana, Vrazov trg 2, 1000, Ljubljana, Slovenia
| | - José L Toca-Herrera
- Institute for Biophysics, Department of Nanobiotechnology, University of Natural Resources and Life Sciences Vienna, Muthgasse 11, A-1190, Vienna, Austria.
| |
Collapse
|
147
|
B A, Rao S, Pandya HJ. Engineering approaches for characterizing soft tissue mechanical properties: A review. Clin Biomech (Bristol, Avon) 2019; 69:127-140. [PMID: 31344655 DOI: 10.1016/j.clinbiomech.2019.07.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 03/14/2019] [Accepted: 07/15/2019] [Indexed: 02/07/2023]
Abstract
From cancer diagnosis to detailed characterization of arterial wall biomechanics, the elastic property of tissues is widely studied as an early sign of disease onset. The fibrous structural features of tissues are a direct measure of its health and functionality. Alterations in the structural features of tissues are often manifested as local stiffening and are early signs for diagnosing a disease. These elastic properties are measured ex vivo in conventional mechanical testing regimes, however, the heterogeneous microstructure of tissues can be accurately resolved over relatively smaller length scales with enhanced spatial resolution using techniques such as micro-indentation, microelectromechanical (MEMS) based cantilever sensors and optical catheters which also facilitate in vivo assessment of mechanical properties. In this review, we describe several probing strategies (qualitative and quantitative) based on the spatial scale of mechanical assessment and also discuss the potential use of machine learning techniques to compute the mechanical properties of soft tissues. This work details state of the art advancement in probing strategies, associated challenges toward quantitative characterization of tissue biomechanics both from an engineering and clinical standpoint.
Collapse
Affiliation(s)
- Alekya B
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore 12, India
| | - Sanjay Rao
- Department of Pediatric Surgery, Mazumdar Shaw Multispecialty Hospital, Narayana Health, Bangalore 99, India
| | - Hardik J Pandya
- Biomedical and Electronic (10(-6)-10(-9)) Engineering Systems Laboratory, Department of Electronic Systems Engineering, Indian Institute of Science, Bangalore 12, India.
| |
Collapse
|
148
|
Mandriota N, Friedsam C, Jones-Molina JA, Tatem KV, Ingber DE, Sahin O. Cellular nanoscale stiffness patterns governed by intracellular forces. NATURE MATERIALS 2019; 18:1071-1077. [PMID: 31209386 PMCID: PMC6754298 DOI: 10.1038/s41563-019-0391-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 04/30/2019] [Indexed: 05/24/2023]
Abstract
Cell stiffness measurements have led to insights into various physiological and pathological processes1,2. Although many cellular behaviours are influenced by intracellular mechanical forces3-6 that also alter the material properties of the cell, the precise mechanistic relationship between intracellular forces and cell stiffness remains unclear. Here we develop a cell mechanical imaging platform with high spatial resolution that reveals the existence of nanoscale stiffness patterns governed by intracellular forces. On the basis of these findings, we develop and validate a cellular mechanical model that quantitatively relates cell stiffness to intracellular forces. This allows us to determine the magnitude of tension within actin bundles, cell cortex and plasma membrane from the cell stiffness patterns across individual cells. These results expand our knowledge on the mechanical interaction between cells and their environments, and offer an alternative approach to determine physiologically relevant intracellular forces from high-resolution cell stiffness images.
Collapse
Affiliation(s)
- Nicola Mandriota
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Claudia Friedsam
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | | | - Kathleen V Tatem
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Department of Physics, Columbia University, New York, NY, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Vascular Biology Program and Departments of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ozgur Sahin
- Department of Biological Sciences, Columbia University, New York, NY, USA.
- Department of Physics, Columbia University, New York, NY, USA.
| |
Collapse
|
149
|
Determination of the Elastic Moduli of a Single Cell Cultured on a Rigid Support by Force Microscopy. Biophys J 2019; 114:2923-2932. [PMID: 29925028 DOI: 10.1016/j.bpj.2018.05.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/08/2018] [Accepted: 05/09/2018] [Indexed: 11/22/2022] Open
Abstract
The elastic response of a living cell is affected by its physiological state. This property provides mechanical fingerprints of a cell's dysfunctionality. The softness (kilopascal range) and thickness (2-15 μm) of mammalian cells imply that the force exerted by the probe might be affected by the stiffness of the solid support. This observation makes infinite sample thickness models unsuitable to describe quantitatively the forces and deformations on a cell. Here, we report a general theory to determine the true Young's moduli of a single cell from a force-indentation curve. Analytical expressions are deduced for common geometries such as flat punches, paraboloids, cones, needles, and nanowires. For a given cell and indentation, the influence of the solid support on the measurements is reduced by using sharp and high aspect ratio tips. The theory is validated by finite element simulations.
Collapse
|
150
|
Et-Thakafy O, Guyomarc'h F, Lopez C. Young modulus of supported lipid membranes containing milk sphingomyelin in the gel, fluid or liquid-ordered phase, determined using AFM force spectroscopy. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:1523-1532. [DOI: 10.1016/j.bbamem.2019.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/12/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022]
|