201
|
Li N, Wang C, Sun S, Zhang C, Lü D, Chen Q, Long M. Microgravity-Induced Alterations of Inflammation-Related Mechanotransduction in Endothelial Cells on Board SJ-10 Satellite. Front Physiol 2018; 9:1025. [PMID: 30108515 PMCID: PMC6079262 DOI: 10.3389/fphys.2018.01025] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 07/11/2018] [Indexed: 12/19/2022] Open
Abstract
Endothelial cells (ECs) are mechanosensitive cells undergoing morphological and functional changes in space. Ground-based study has provided a body of evidences about how ECs can respond to the effect of simulated microgravity, however, these results need to be confirmed by spaceflight experiments in real microgravity. In this work, we cultured EA.hy926 ECs on board the SJ-10 Recoverable Scientific Satellite for 3 and 10 days, and analyzed the effects of space microgravity on the ECs. Space microgravity suppressed the glucose metabolism, modulated the expression of cellular adhesive molecules such as ICAM-1, VCAM-1, and CD44, and depressed the pro-angiogenesis and pro-inflammation cytokine secretion. Meanwhile, it also induced the depolymerization of actin filaments and microtubules, promoted the vimentin accumulation, restrained the collagen I and fibronectin deposition, regulated the mechanotransduction through focal adhesion kinase and Rho GTPases, and enhanced the exosome-mediated mRNA transfer. Unlike the effect of simulated microgravity, neither three-dimensional growth nor enhanced nitric oxide production was observed in our experimental settings. This work furthers the understandings in the effects and mechanisms of space microgravity on ECs, and provides useful information for future spaceflight experimental design.
Collapse
Affiliation(s)
- Ning Li
- Key Laboratory of Microgravity - National Microgravity Laboratory, Center of Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chengzhi Wang
- Key Laboratory of Microgravity - National Microgravity Laboratory, Center of Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shujin Sun
- Key Laboratory of Microgravity - National Microgravity Laboratory, Center of Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chen Zhang
- Key Laboratory of Microgravity - National Microgravity Laboratory, Center of Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Dongyuan Lü
- Key Laboratory of Microgravity - National Microgravity Laboratory, Center of Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qin Chen
- Key Laboratory of Microgravity - National Microgravity Laboratory, Center of Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Mian Long
- Key Laboratory of Microgravity - National Microgravity Laboratory, Center of Biomechanics and Bioengineering, Beijing Key Laboratory of Engineered Construction and Mechanobiology, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China.,School of Engineering Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
202
|
Kim R, Wang Y, Hwang SHJ, Attayek PJ, Smiddy NM, Reed MI, Sims CE, Allbritton NL. Formation of arrays of planar, murine, intestinal crypts possessing a stem/proliferative cell compartment and differentiated cell zone. LAB ON A CHIP 2018; 18:2202-2213. [PMID: 29944153 PMCID: PMC6337012 DOI: 10.1039/c8lc00332g] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A simple, in vitro intestinal model recapitulating key aspects of crypt architecture and physiology would facilitate our understanding the impact of drugs, foods and microbial metabolites on the intestine. To address the limitations of previously reported intestinal in vitro platforms, we developed a planar crypt array that replicated the spatial segregation and physiologic responses of primary mouse intestinal epithelial cells in the large intestine. Collagen was coated across an impermeable film possessing an array of microholes creating two regions of distinct stiffness and porosity (above and outside the microholes). Primary mouse colon epithelial cells formed a continuous monolayer across the array with a proliferative cell zone above the microholes and a nonproliferative or differentiated cell region distant from the microholes. Formation of a chemical gradient of growth factors across the array yielded a more complete or in vivo-like cell segregation of proliferative and differentiated cells with cell migration outward from the proliferative cell zone into the differentiated zone to replace apoptotic dying cells much as occurs in vivo. Short chain fatty acids (microbial metabolites) applied to the luminal surface of the crypt array significantly impacted the proliferation and differentiation of the cells replicating the known in vivo effects of these fatty acids. Importantly this planar crypt array was readily fabricated and maintained, easily imaged with properties quantified by microscopy, and compatible with reagent addition to either the luminal or basal fluid reservoirs. The ability to observe simultaneously stem/proliferative and differentiated cell behavior and movement between these two compartments in response to drugs, toxins, inflammatory mediators or microbial metabolites will be of widespread utility.
Collapse
Affiliation(s)
- Raehyun Kim
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, and North Carolina State University, Raleigh, North Carolina, USA.
| | | | | | | | | | | | | | | |
Collapse
|
203
|
LaCroix AS, Lynch AD, Berginski ME, Hoffman BD. Tunable molecular tension sensors reveal extension-based control of vinculin loading. eLife 2018; 7:33927. [PMID: 30024378 PMCID: PMC6053308 DOI: 10.7554/elife.33927] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 06/03/2018] [Indexed: 01/03/2023] Open
Abstract
Molecular tension sensors have contributed to a growing understanding of mechanobiology. However, the limited dynamic range and inability to specify the mechanical sensitivity of these sensors has hindered their widespread use in diverse contexts. Here, we systematically examine the components of tension sensors that can be altered to improve their functionality. Guided by the development of a first principles model describing the mechanical behavior of these sensors, we create a collection of sensors that exhibit predictable sensitivities and significantly improved performance in cellulo. Utilized in the context of vinculin mechanobiology, a trio of these new biosensors with distinct force- and extension-sensitivities reveal that an extension-based control paradigm regulates vinculin loading in a variety of mechanical contexts. To enable the rational design of molecular tension sensors appropriate for diverse applications, we predict the mechanical behavior, in terms of force and extension, of additional 1020 distinct designs. Cells must sense signals from their surroundings to play their roles within the body. These signals can be biochemical, such as growth-promoting substances, or mechanical, for example the stiffness or softness of the environment. Mechanical signals can be detected by load-bearing proteins, which stretch like tiny springs in response to forces. In animals, these proteins span the membrane separating the interior of the cell from the exterior. Externally, the proteins attach to structures around the cell; internally, they connect to the machinery that both generates forces and allows cells to respond to signals from outside. As such, load-bearing proteins form a direct mechanical link between cell and environment. Scientists use tools called molecular tension sensors to measure how much a load-bearing protein stretches in response to changes, and the force that is being applied to it. However, just like any other type of scale, these sensors only work over a certain range, which happens to be limited. This means that, for example, they cannot measure forces in tissues that are too soft (like the brain), or too stiff (such as bones). New sensors that can assess forces in these contexts are therefore needed, but so far research in this area has been slow due to a reliance on ‘trial-and-error’ approaches. Here, LaCroix et al. developed a new method to predict the sensitivity of molecular tension sensors inside cells. This was accomplished by examining several existing sensors, and identifying which components could be altered to change the properties of the sensors. Then, this information was used to create a computer model that could predict how new sensors would behave, and which range of forces they could measure. Finally, the sensors designed following this method were tested in mouse cells grown in the laboratory, and they worked better than their predecessors. The next step was for LaCroix et al. to use a trio of new sensors with different sensitivities to study the load-bearing protein vinculin in mouse cells. The goal was to figure out exactly how cells manage their load-bearing proteins. Indeed, it was widely assumed that a cell acts on a load-bearing protein by applying a force on it. In response, the protein would stretch by a certain amount, which can change depending on its properties – a ‘stiffer’ protein would stretch less. Unexpectedly, the new sensors showed that cells instead manipulate how much vinculin stretches, applying varying forces to achieve the same length of the protein in different environments. Improved molecular tension sensors will give scientists a better insight into how cells respond to their mechanical environment, which could help to direct cell behavior in tissues engineered in the laboratory. This knowledge is also directly relevant to human health, as the mechanical properties of many tissues change during disease, such as tumors stiffening during cancer.
Collapse
Affiliation(s)
- Andrew S LaCroix
- Department of Biomedical Engineering, Duke University, Durham, United States
| | - Andrew D Lynch
- Department of Biomedical Engineering, Duke University, Durham, United States
| | - Matthew E Berginski
- Department of Biomedical Engineering, Duke University, Durham, United States
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, United States
| |
Collapse
|
204
|
Giannini M, Primerano C, Berger L, Giannaccini M, Wang Z, Landi E, Cuschieri A, Dente L, Signore G, Raffa V. Nano-topography: Quicksand for cell cycle progression? NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2656-2665. [PMID: 30010000 DOI: 10.1016/j.nano.2018.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/26/2018] [Accepted: 07/01/2018] [Indexed: 01/01/2023]
Abstract
The 3-D spatial and mechanical features of nano-topography can create alternative environments, which influence cellular response. In this paper, murine fibroblast cells were grown on surfaces characterized by protruding nanotubes. Cells cultured on such nano-structured surface exhibit stronger cellular adhesion compared to control groups, but despite the fact that stronger adhesion is generally believed to promote cell cycle progression, the time cells spend in G1 phase is doubled. This apparent contradiction is solved by confocal microscopy analysis, which shows that the nano-topography inhibits actin stress fiber formation. In turn, this impairs RhoA activation, which is required to suppress the inhibition of cell cycle progression imposed by p21/p27. This finding suggests that the generation of stress fibers, required to impose the homeostatic intracellular tension, rather than cell adhesion/spreading is the limiting factor for cell cycle progression. Indeed, nano-topography could represent a unique tool to inhibit proliferation in adherent well-spread cells.
Collapse
Affiliation(s)
| | | | - Liron Berger
- Department of Biology, Università di Pisa, Pisa, Italy.
| | | | - Zhigang Wang
- Institute for Medical Science and Technology, University of Dundee, Dundee, United Kingdom.
| | - Elena Landi
- Department of Biology, Università di Pisa, Pisa, Italy.
| | - Alfred Cuschieri
- Institute for Medical Science and Technology, University of Dundee, Dundee, United Kingdom.
| | - Luciana Dente
- Department of Biology, Università di Pisa, Pisa, Italy.
| | - Giovanni Signore
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Pisa, Italy; NEST, Scuola Normale Superiore, and Istituto Nanoscienze-CNR, Pisa, Italy.
| | | |
Collapse
|
205
|
Shah JS, Sabouni R, Cayton Vaught KC, Owen CM, Albertini DF, Segars JH. Biomechanics and mechanical signaling in the ovary: a systematic review. J Assist Reprod Genet 2018; 35:1135-1148. [PMID: 29691711 PMCID: PMC6063820 DOI: 10.1007/s10815-018-1180-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/05/2018] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Mammalian oogenesis and folliculogenesis share a dynamic connection that is critical for gamete development. For maintenance of quiescence or follicular activation, follicles must respond to soluble signals (growth factors and hormones) and physical stresses, including mechanical forces and osmotic shifts. Likewise, mechanical processes are involved in cortical tension and cell polarity in oocytes. Our objective was to examine the contribution and influence of biomechanical signaling in female mammalian gametogenesis. METHODS We performed a systematic review to assess and summarize the effects of mechanical signaling and mechanotransduction in oocyte maturation and folliculogenesis and to explore possible clinical applications. The review identified 2568 publications of which 122 met the inclusion criteria. RESULTS The integration of mechanical and cell signaling pathways in gametogenesis is complex. Follicular activation or quiescence are influenced by mechanical signaling through the Hippo and Akt pathways involving the yes-associated protein (YAP), transcriptional coactivator with PDZ-binding motif (TAZ), phosphatase and tensin homolog deleted from chromosome 10 (PTEN) gene, the mammalian target of rapamycin (mTOR), and forkhead box O3 (FOXO3) gene. CONCLUSIONS There is overwhelming evidence that mechanical signaling plays a crucial role in development of the ovary, follicle, and oocyte throughout gametogenesis. Emerging data suggest the complexities of mechanotransduction and the biomechanics of oocytes and follicles are integral to understanding of primary ovarian insufficiency, ovarian aging, polycystic ovary syndrome, and applications of fertility preservation.
Collapse
Affiliation(s)
- Jaimin S Shah
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Texas at Houston Health Science Center, Houston, TX, USA
| | - Reem Sabouni
- Jones Institute for Reproductive Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Kamaria C Cayton Vaught
- Howard W. and Georgeanna Seegar Jones Division of Reproductive Sciences and Women's Health Research, Baltimore, MD, USA
| | - Carter M Owen
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | | | - James H Segars
- Howard W. and Georgeanna Seegar Jones Division of Reproductive Sciences and Women's Health Research, Baltimore, MD, USA.
- Gynecology and Obstetrics, 720 Rutland Avenue/Ross 624, Baltimore, MD, 21205, USA.
| |
Collapse
|
206
|
Allam AH, Charnley M, Russell SM. Context-Specific Mechanisms of Cell Polarity Regulation. J Mol Biol 2018; 430:3457-3471. [PMID: 29886017 DOI: 10.1016/j.jmb.2018.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/04/2018] [Accepted: 06/04/2018] [Indexed: 12/31/2022]
Abstract
Cell polarity is an essential process shared by almost all animal tissues. Moreover, cell polarity enables cells to sense and respond to the cues provided by the neighboring cells and the surrounding microenvironment. These responses play a critical role in regulating key physiological processes, including cell migration, proliferation, differentiation, vesicle trafficking and immune responses. The polarity protein complexes regulating these interactions are highly evolutionarily conserved between vertebrates and invertebrates. Interestingly, these polarity complexes interact with each other and key signaling pathways in a cell-polarity context-dependent manner. However, the exact mechanisms by which these interactions take place are poorly understood. In this review, we will focus on the roles of the key polarity complexes SCRIB, PAR and Crumbs in regulating different forms of cell polarity, including epithelial cell polarity, cell migration, asymmetric cell division and the T-cell immunological synapse assembly and signaling.
Collapse
Affiliation(s)
- Amr H Allam
- Centre for Micro-Photonics, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Australia; Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Australia.
| | - Mirren Charnley
- Centre for Micro-Photonics, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Australia; Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Australia; Biointerface Engineering Group, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC, Australia.
| | - Sarah M Russell
- Centre for Micro-Photonics, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Australia; Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Parkville, Australia; Department of Pathology, The University of Melbourne, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Australia.
| |
Collapse
|
207
|
McCarthy JB, El-Ashry D, Turley EA. Hyaluronan, Cancer-Associated Fibroblasts and the Tumor Microenvironment in Malignant Progression. Front Cell Dev Biol 2018; 6:48. [PMID: 29868579 PMCID: PMC5951929 DOI: 10.3389/fcell.2018.00048] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the roles of CAFs in forming a “cancerized” fibrotic stroma favorable to tumor initiation and dissemination, in particular highlighting the functions of the extracellular matrix component hyaluronan (HA) in these processes. The structural complexity of the tumor and its host microenvironment is now well appreciated to be an important contributing factor to malignant progression and resistance-to-therapy. There are multiple components of this complexity, which include an extensive remodeling of the extracellular matrix (ECM) and associated biomechanical changes in tumor stroma. Tumor stroma is often fibrotic and rich in fibrillar type I collagen and hyaluronan (HA). Cancer-associated fibroblasts (CAFs) are a major source of this fibrotic ECM. CAFs organize collagen fibrils and these biomechanical alterations provide highways for invading carcinoma cells either under the guidance of CAFs or following their epithelial to mesenchymal transition (EMT). The increased HA metabolism of a tumor microenvironment instructs carcinoma initiation and dissemination by performing multiple functions. The key effects of HA reviewed here are its role in activating CAFs in pre-malignant and malignant stroma, and facilitating invasion by promoting motility of both CAFs and tumor cells, thus facilitating their invasion. Circulating CAFs (cCAFs) also form heterotypic clusters with circulating tumor cells (CTC), which are considered to be pre-cursors of metastatic colonies. cCAFs are likely required for extravasation of tumors cells and to form a metastatic niche suitable for new tumor colony growth. Therapeutic interventions designed to target both HA and CAFs in order to limit tumor spread and increase response to current therapies are discussed.
Collapse
Affiliation(s)
- James B McCarthy
- Department of Laboratory Medicine and Pathology, Masonic Comprehensive Cancer Center, Minneapolis, MN, United States
| | - Dorraya El-Ashry
- Department of Laboratory Medicine and Pathology, Masonic Comprehensive Cancer Center, Minneapolis, MN, United States
| | - Eva A Turley
- London Regional Cancer Program, Department of Oncology, Biochemistry and Surgery, Schulich School of Medicine and Dentistry, Lawson Health Research Institute, Western University, London, ON, Canada
| |
Collapse
|
208
|
Choi S, Friedrichs J, Song YH, Werner C, Estroff LA, Fischbach C. Intrafibrillar, bone-mimetic collagen mineralization regulates breast cancer cell adhesion and migration. Biomaterials 2018; 198:95-106. [PMID: 29759731 DOI: 10.1016/j.biomaterials.2018.05.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/27/2018] [Accepted: 05/01/2018] [Indexed: 10/17/2022]
Abstract
Bone metastasis is a leading cause of death in patients with breast cancer, but the underlying mechanisms are poorly understood. While much work focuses on the molecular and cellular events that drive breast cancer bone metastasis, it is mostly unclear what role bone extracellular matrix (ECM) properties play in this process. Bone ECM primarily consists of mineralized collagen fibrils, which are composed of non-stoichiometric carbonated apatite (HA) and collagen type I. Reduced bone mineral content is epidemiologically linked with increased risk of bone metastasis. Yet elucidating the potential functional impact of collagen mineralization on breast cancer cells has remained challenging because of a lack of model systems that allow studying tumor cell behavior as a function of physiological, intrafibrillar collagen mineralization. Here, we have developed cell culture substrates composed of mineralized collagen type I fibrils using a polymer-induced liquid-precursor (PILP) process. Intrafibrillar HA decreased breast cancer cell adhesion forces and accordingly reduced collagen fiber alignment relative to cells cultured on control collagen. The resulting mineral-mediated changes in collagen network characteristics and mechanosignaling correlated with increased cell motility, but inhibited directed migration of breast cancer cells. These results suggest that physiological mineralization of collagen fibrils reduces tumor cell adhesion with potential functional consequences on skeletal homing of disseminated tumor cells in early stages of breast cancer metastasis.
Collapse
Affiliation(s)
- Siyoung Choi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jens Friedrichs
- Institute of Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany
| | - Young Hye Song
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Carsten Werner
- Institute of Biofunctional Polymer Materials, Leibniz Institute of Polymer Research Dresden, 01069 Dresden, Germany
| | - Lara A Estroff
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
209
|
Combined VEGF/PDGF inhibition using axitinib induces αSMA expression and a pro-fibrotic phenotype in human pericytes. Graefes Arch Clin Exp Ophthalmol 2018; 256:1141-1149. [PMID: 29721663 DOI: 10.1007/s00417-018-3987-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 03/06/2018] [Accepted: 04/11/2018] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Large trials on anti-VEGF/PDGF (vascular endothelial/platelet-derived growth factor) combination therapy have been established to improve management of neovascular activity in age-related macular degeneration. Targeting pericytes, PDGF is thought to induce vessel regression and reduce fibrovascular scarring. The fate of pericytes exposed to anti-VEGF/PDGF combination therapy is not clear. Therefore, this study was designed to study the influence of anti-VEGF/PDGF on pericyte phenotype and cellular behavior. METHODS Human pericytes from placenta (hPC-PL) were treated with axitinib, a tyrosine kinase inhibitor targeting VEGFR1-3 and PDGFR. Toxic effects were excluded using live/dead staining. Phenotypic changes were evaluated using phalloidin staining for actin cytoskeleton and the expression of stress fibers. MRNA and protein expression levels of α-smooth muscle actin (αSMA) as a marker of proto-myofibroblastic transition were evaluated with real-time PCR and Western blotting. Influences of fibrotic cellular mechanisms were evaluated with a scratch wound migration and a collagen gel contraction assay. RESULTS Treatment with 0.5, 1, and 2.5 μg/ml axitinib strongly induced a proto-myofibroblast-like actin cytoskeleton with a marked increase in stress fibers. Quantitative real-time PCR and Western blotting revealed these changes to be linked to dose-dependent increases in αSMA mRNA and protein expression. However, fibrotic cellular mechanisms were significantly reduced in the presence of axitinib (scratch wound closure: up to - 78.4%, collagen gel contraction: up to - 37.4%). CONCLUSIONS Combined anti-VEGF/PDGF inhibition seems to induce a proto-myofibroblast-like phenotype in human pericytes in vitro, but reduce profibrotic cellular mechanisms due to prolonged anti-PDGF inhibition.
Collapse
|
210
|
Spyridopoulou K, Makridis A, Maniotis N, Karypidou N, Myrovali E, Samaras T, Angelakeris M, Chlichlia K, Kalogirou O. Effect of low frequency magnetic fields on the growth of MNP-treated HT29 colon cancer cells. NANOTECHNOLOGY 2018; 29:175101. [PMID: 29498936 DOI: 10.1088/1361-6528/aaaea9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Recent investigations have attempted to understand and exploit the impact of magnetic field-actuated internalized magnetic nanoparticles (MNPs) on the proliferation rate of cancer cells. Due to the complexity of the parameters governing magnetic field-exposure though, individual studies to date have raised contradictory results. In our approach we performed a comparative analysis of key parameters related to the cell exposure of cancer cells to magnetic field-actuated MNPs, and to the magnetic field, in order to better understand the factors affecting cellular responses to magnetic field-stimulated MNPs. We used magnetite MNPs with a hydrodynamic diameter of 100 nm and studied the proliferation rate of MNPs-treated versus untreated HT29 human colon cancer cells, exposed to either static or alternating low frequency magnetic fields with varying intensity (40-200 mT), frequency (0-8 Hz) and field gradient. All three parameters, field intensity, frequency, and field gradient affected the growth rate of cells, with or without internalized MNPs, as compared to control MNPs-untreated and magnetic field-untreated cells. We observed that the growth inhibitory effects induced by static and rotating magnetic fields were enhanced by pre-treating the cells with MNPs, while the growth promoting effects observed in alternating field-treated cells were weakened by MNPs. Compared to static, rotating magnetic fields of the same intensity induced a similar extend of cell growth inhibition, while alternating fields of varying intensity (70 or 100 mT) and frequency (0, 4 or 8 Hz) induced cell proliferation in a frequency-dependent manner. These results, highlighting the diverse effects of mode, intensity, and frequency of the magnetic field on cell growth, indicate that consistent and reproducible results can be achieved by controlling the complexity of the exposure of biological samples to MNPs and external magnetic fields, through monitoring crucial experimental parameters. We demonstrate that further research focusing on the accurate manipulation of the aforementioned magnetic field exposure parameters could lead to the development of successful non-invasive therapeutic anticancer approaches.
Collapse
Affiliation(s)
- K Spyridopoulou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | | | | | | | | | | | | | | | | |
Collapse
|
211
|
Narkhede AA, Crenshaw JH, Manning RM, Rao SS. The influence of matrix stiffness on the behavior of brain metastatic breast cancer cells in a biomimetic hyaluronic acid hydrogel platform. J Biomed Mater Res A 2018; 106:1832-1841. [DOI: 10.1002/jbm.a.36379] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 02/04/2018] [Accepted: 02/15/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Akshay A. Narkhede
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosa Alabama
| | - James H. Crenshaw
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosa Alabama
| | - Riley M. Manning
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosa Alabama
| | - Shreyas S. Rao
- Department of Chemical and Biological EngineeringThe University of AlabamaTuscaloosa Alabama
| |
Collapse
|
212
|
Deng W, Wang Y, Zhao S, Zhang Y, Chen Y, Zhao X, Liu L, Sun S, Zhang L, Ye B, Du J. MICAL1 facilitates breast cancer cell proliferation via ROS-sensitive ERK/cyclin D pathway. J Cell Mol Med 2018. [PMID: 29524295 PMCID: PMC5980113 DOI: 10.1111/jcmm.13588] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Molecule interacting with CasL 1 (MICAL1) is a multidomain flavoprotein mono-oxygenase that strongly involves in cytoskeleton dynamics and cell oxidoreduction metabolism. Recently, results from our laboratory have shown that MICAL1 modulates reactive oxygen species (ROS) production, and the latter then activates phosphatidyl inositol 3-kinase (PI3K)/protein kinase B (Akt) signalling pathway which regulates breast cancer cell invasion. Herein, we performed this study to assess the involvement of MICAL1 in breast cancer cell proliferation and to explore the potential molecular mechanism. We noticed that depletion of MICAL1 markedly reduced cell proliferation in breast cancer cell line MCF-7 and T47D. This effect of MICAL1 on proliferation was independent of wnt/β-catenin and NF-κB pathways. Interestingly, depletion of MICAL1 significantly inhibited ROS production, decreased p-ERK expression and unfavourable for proliferative phenotype of breast cancer cells. Likewise, MICAL1 overexpression increased p-ERK level as well as p-ERK nucleus translocation. Moreover, we investigated the effect of MICAL1 on cell cycle-related proteins. MICAL1 positively regulated CDK4 and cyclin D expression, but not CDK2, CDK6, cyclin A and cyclin E. In addition, more expression of CDK4 and cyclin D by MICAL1 overexpression was blocked by PI3K/Akt inhibitor LY294002. LY294002 treatment also attenuated the increase in the p-ERK level in MICAL1-overexpressed breast cancer cells. Together, our results suggest that MICAL1 exhibits its effect on proliferation via maintaining cyclin D expression through ROS-sensitive PI3K/Akt/ERK signalling in breast cancer cells.
Collapse
Affiliation(s)
- Wenjie Deng
- Department of Physiology, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yueyuan Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Shuo Zhao
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Yujie Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yan Chen
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Xuyang Zhao
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Lei Liu
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Shixiu Sun
- Department of Physiology, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Lin Zhang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.,Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Bixing Ye
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Jun Du
- Department of Physiology, Nanjing Medical University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center For Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
213
|
Northcott JM, Dean IS, Mouw JK, Weaver VM. Feeling Stress: The Mechanics of Cancer Progression and Aggression. Front Cell Dev Biol 2018. [PMID: 29541636 PMCID: PMC5835517 DOI: 10.3389/fcell.2018.00017] [Citation(s) in RCA: 274] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The tumor microenvironment is a dynamic landscape in which the physical and mechanical properties evolve dramatically throughout cancer progression. These changes are driven by enhanced tumor cell contractility and expansion of the growing tumor mass, as well as through alterations to the material properties of the surrounding extracellular matrix (ECM). Consequently, tumor cells are exposed to a number of different mechanical inputs including cell–cell and cell-ECM tension, compression stress, interstitial fluid pressure and shear stress. Oncogenes engage signaling pathways that are activated in response to mechanical stress, thereby reworking the cell's intrinsic response to exogenous mechanical stimuli, enhancing intracellular tension via elevated actomyosin contraction, and influencing ECM stiffness and tissue morphology. In addition to altering their intracellular tension and remodeling the microenvironment, cells actively respond to these mechanical perturbations phenotypically through modification of gene expression. Herein, we present a description of the physical changes that promote tumor progression and aggression, discuss their interrelationship and highlight emerging therapeutic strategies to alleviate the mechanical stresses driving cancer to malignancy.
Collapse
Affiliation(s)
- Josette M Northcott
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States
| | - Ivory S Dean
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States
| | - Janna K Mouw
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, CA, United States.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, United States.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States.,UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
214
|
Bao Y, Guo H, Lu Y, Feng W, Sun X, Tang C, Wang X, Shen M. Blocking hepatic metastases of colon cancer cells using an shRNA against Rac1 delivered by activatable cell-penetrating peptide. Oncotarget 2018; 7:77183-77195. [PMID: 27791203 PMCID: PMC5363579 DOI: 10.18632/oncotarget.12854] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 10/14/2016] [Indexed: 12/27/2022] Open
Abstract
Hepatic metastasis is one of the critical progressions of colon cancer. Blocking this process is key to prolonging survival time in cancer patients. Studies on activatable cell-penetrating peptides (dtACPPs) have demonstrated their potential as gene carriers. It showed high tumor cell-targeting specificity and transfection efficiency and low cytotoxicity in the in vitro settings of drug delivery. However, using this system to silence target genes to inhibit metastasis in colorectal cancer cells has not been widely reported and requires further investigation. In this study, we observed that expression of Rac1, a key molecule for cytoskeletal reorganization, was higher in hepatic metastatic tumor tissue compared with prime colon cancer tissue and that patients with high Rac1-expressing colon cancer showed shorter survival time. Base on these findings, we created dtACPP-PEG-DGL (dtACPPD)/shRac1 nanoparticles and demonstrated that they downregulated Rac1 expression in colon cancer cells. Moreover, we observed inhibitory effects on migration, invasion and adhesion in HCT116 colorectal cancer cells in vitro, and our results showed that Rac1 regulated colon cancer cell matrix adhesion through the regulation of cytofilament dynamics. Moreover, mechanically, repression of Rac1 inhibiting cells migration and invasion by enhancing cell to cell adhesion and reducing cell to extracellular matrix adhesion. Furthermore, when atCDPPD/shRac1 nanoparticles were administered intravenously to a HCT116 xenograft model, significant tumor metastasis to the liver was inhibited. Our results suggest that atCDPP/shRac1 nanoparticles may enable the blockade of hepatic metastasis in colon cancer.
Collapse
Affiliation(s)
- Ying Bao
- Department of Surgery, First Affiliated Hospital, Huzhou University, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Huihui Guo
- Department of Surgery, First Affiliated Hospital, Huzhou University, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Yongliang Lu
- Department of Medicine, Huzhou University, Huzhou, 313000, China
| | - Wenming Feng
- Department of Surgery, First Affiliated Hospital, Huzhou University, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Xinrong Sun
- Department of Surgery, First Affiliated Hospital, Huzhou University, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Chengwu Tang
- Department of Surgery, First Affiliated Hospital, Huzhou University, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Xiang Wang
- Department of Surgery, First Affiliated Hospital, Huzhou University, The First People's Hospital of Huzhou, Huzhou, 313000, China
| | - Mo Shen
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University,Wenzhou, 325000, China
| |
Collapse
|
215
|
Berger SL, Leo-Macias A, Yuen S, Khatri L, Pfennig S, Zhang Y, Agullo-Pascual E, Caillol G, Zhu MS, Rothenberg E, Melendez-Vasquez CV, Delmar M, Leterrier C, Salzer JL. Localized Myosin II Activity Regulates Assembly and Plasticity of the Axon Initial Segment. Neuron 2018; 97:555-570.e6. [PMID: 29395909 PMCID: PMC5805619 DOI: 10.1016/j.neuron.2017.12.039] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 08/24/2017] [Accepted: 12/22/2017] [Indexed: 01/08/2023]
Abstract
The axon initial segment (AIS) is the site of action potential generation and a locus of activity-dependent homeostatic plasticity. A multimeric complex of sodium channels, linked via a cytoskeletal scaffold of ankyrin G and beta IV spectrin to submembranous actin rings, mediates these functions. The mechanisms that specify the AIS complex to the proximal axon and underlie its plasticity remain poorly understood. Here we show phosphorylated myosin light chain (pMLC), an activator of contractile myosin II, is highly enriched in the assembling and mature AIS, where it associates with actin rings. MLC phosphorylation and myosin II contractile activity are required for AIS assembly, and they regulate the distribution of AIS components along the axon. pMLC is rapidly lost during depolarization, destabilizing actin and thereby providing a mechanism for activity-dependent structural plasticity of the AIS. Together, these results identify pMLC/myosin II activity as a common link between AIS assembly and plasticity.
Collapse
Affiliation(s)
- Stephen L Berger
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - Stephanie Yuen
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Latika Khatri
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Sylvia Pfennig
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | - Yanqing Zhang
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - Ghislaine Caillol
- Aix Marseille Université, CNRS, INP UMR7051, 13344 Cedex 15, Marseille, France
| | - Min-Sheng Zhu
- Model Animal Research Center and MOE Key Laboratory of Model Animal and Disease Study, Nanjing University, Nanjing 210061, China
| | - Eli Rothenberg
- Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, NY 10016, USA
| | - Carmen V Melendez-Vasquez
- Department of Biological Sciences, Hunter College, New York, NY 10065, USA; Department of Molecular, Cellular, and Developmental Biology, The Graduate Center, The City University of New York, NY 10016, USA
| | - Mario Delmar
- Division of Cardiology, NYU School of Medicine, New York, NY 10016, USA
| | | | - James L Salzer
- Neuroscience Institute and Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
216
|
Barui A, Chowdhury F, Pandit A, Datta P. Rerouting mesenchymal stem cell trajectory towards epithelial lineage by engineering cellular niche. Biomaterials 2018; 156:28-44. [DOI: 10.1016/j.biomaterials.2017.11.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 10/22/2017] [Accepted: 11/21/2017] [Indexed: 02/06/2023]
|
217
|
Goreczny GJ, Forsythe IJ, Turner CE. Hic-5 regulates fibrillar adhesion formation to control tumor extracellular matrix remodeling through interaction with tensin1. Oncogene 2018; 37:1699-1713. [PMID: 29348458 PMCID: PMC5876083 DOI: 10.1038/s41388-017-0074-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 11/02/2017] [Accepted: 11/19/2017] [Indexed: 01/26/2023]
Abstract
The linearization of the stromal extracellular matrix (ECM) by cancer associated fibroblasts (CAFs) facilitates tumor cell growth and metastasis. However, the mechanism by which the ECM is remodeled is not fully understood. Hic-5 (TGFβ1i1), a focal adhesion scaffold protein, has previously been reported to be crucial for stromal ECM deposition and remodeling in vivo. Herein we show that CAFs lacking Hic-5 exhibit a significant reduction in the ability to form fibrillar adhesions, a specialized form of focal adhesion that promote fibronectin fibrillogenesis. Hic-5 was found to promote fibrillar adhesion formation through a newly characterized interaction with tensin1. Furthermore, Src dependent phosphorylation of Hic-5 facilitated the interaction with tensin1 to prevent β1 integrin internalization and trafficking to the lysosome. The interaction between Hic-5 and tensin1 was mechanosensitive, promoting fibrillar adhesion formation and fibronectin fibrillogenesis in a rigidity dependent fashion. Importantly, this Src dependent mechanism was conserved in three-dimensional (3D) ECM environments. Immunohistochemistry of tensin1 showed enrichment in CAFs in vivo, which was abrogated upon deletion of Hic-5. Interestingly, elevated Hic-5 expression correlates with reduced distant metastasis free survival in patients with basal-like, HER2+ and grade 3 tumors. Thus, we have identified Hic-5 as a crucial regulator of ECM remodeling in CAFs by promoting fibrillar adhesion formation through a novel interaction with tensin1.
Collapse
Affiliation(s)
- Gregory J Goreczny
- Department of Cell & Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Ian J Forsythe
- Department of Cell & Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA
| | - Christopher E Turner
- Department of Cell & Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY, USA.
| |
Collapse
|
218
|
Design of Fiber Networks for Studying Metastatic Invasion. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1092:289-318. [DOI: 10.1007/978-3-319-95294-9_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
219
|
Dental-Derived Stem Cells and Their Secretome and Interactions with Bioscaffolds/Biomaterials in Regenerative Medicine: From the In Vitro Research to Translational Applications. Stem Cells Int 2017; 2017:6975251. [PMID: 29445404 PMCID: PMC5763215 DOI: 10.1155/2017/6975251] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 11/20/2017] [Indexed: 01/04/2023] Open
|
220
|
Huang L, Tian M, Wu D, Duscher G, Liaw PK, He W. Surface Mechanoengineering of a Zr-Based Bulk Metallic Glass via Ar-Nanobubble Doping To Probe Cell Sensitivity to Rigid Materials. ACS APPLIED MATERIALS & INTERFACES 2017; 9:43429-43437. [PMID: 29171273 DOI: 10.1021/acsami.7b12663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In this study, a new materials platform, utilizing the amorphous microstructure of bulk metallic glasses (BMGs) and the versatility of ion implantation, was developed for the fundamental investigation of cell responses to substrate-rigidity variations in the gigapascal modulus range, which was previously unattainable with polymeric materials. The surface rigidity of a Zr-Al-Ni-Cu-Y BMG was modulated with low-energy Ar-ion implantation because of the impartment of Ar nanobubbles into the amorphous matrix. Surface softening was achieved due to the formation of nanobubble-doped transitional zones in the Zr-based BMG substrate. Bone-forming cell studies on this newly designed platform demonstrated that mechanical cues, accompanied by the potential effects of other surface properties (i.e., roughness, morphology, and chemistry), contributed to modulating cell behaviors. Cell adhesion and actin filaments were found to be less established on less stiff surfaces, especially on the surface with an elastic modulus of 51 GPa. Cell growth appeared to be affected by surface-mechanical properties. A lower stiffness was generally related to a higher growth rate. Findings in this study broadened our fundamental understanding concerning the mechanosensing of bone cells on stiff substrates. It also suggests that surface mechanoengineering of metallic materials could be a potential strategy to promote osseointegration of such materials for bone-implant applications. Further investigations are proposed to fine-tune the ion implantation variables in order to further distinguish the surface-mechanical effect on bone-forming cell activities from the contributions of other surface properties.
Collapse
Affiliation(s)
- Lu Huang
- Department of Materials Science and Engineering, University of Tennessee , Knoxville, Tennessee 37996, United States
| | - Mengkun Tian
- Department of Chemical and Biomaterials Engineering, University of Tennessee , Knoxville, Tennessee 37996, United States
| | - Dong Wu
- Department of Materials Science and Engineering, University of Tennessee , Knoxville, Tennessee 37996, United States
| | - Gerd Duscher
- Department of Materials Science and Engineering, University of Tennessee , Knoxville, Tennessee 37996, United States
- Materials Science and Technology Division, Oak Ridge National Laboratory , Oak Ridge, Tennessee 37831, United States
| | - Peter K Liaw
- Department of Materials Science and Engineering, University of Tennessee , Knoxville, Tennessee 37996, United States
| | - Wei He
- Department of Materials Science and Engineering, University of Tennessee , Knoxville, Tennessee 37996, United States
| |
Collapse
|
221
|
Bae JC, Lee JJ, Shim JH, Park KH, Lee JS, Bae EB, Choi JW, Huh JB. Development and Assessment of a 3D-Printed Scaffold with rhBMP-2 for an Implant Surgical Guide Stent and Bone Graft Material: A Pilot Animal Study. MATERIALS (BASEL, SWITZERLAND) 2017; 10:E1434. [PMID: 29258172 PMCID: PMC5744369 DOI: 10.3390/ma10121434] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 12/12/2017] [Accepted: 12/14/2017] [Indexed: 01/10/2023]
Abstract
In this study, a new concept of a 3D-printed scaffold was introduced for the accurate placement of an implant and the application of a recombinant human bone morphogenetic protein-2 (rhBMP-2)-loaded bone graft. This preliminary study was conducted using two adult beagles to evaluate the 3D-printed polycaprolactone (PCL)/β-tricalcium phosphate (β-TCP)/bone decellularized extracellular matrix (bdECM) scaffold conjugated with rhBMP-2 for the simultaneous use as an implant surgical guide stent and bone graft material that promotes new bone growth. Teeth were extracted from the mandible of the beagle model and scanned by computed tomography (CT) to fabricate a customized scaffold that would fit the bone defect. After positioning the implant guide scaffold, the implant was placed and rhBMP-2 was injected into the scaffold of the experimental group. The two beagles were sacrificed after three months. The specimen block was obtained and scanned by micro-CT. Histological analysis showed that the control and experimental groups had similar new bone volume (NBV, %) but the experimental group with BMP exhibited a significantly higher bone-to-implant contact ratio (BIC, %). Within the limitations of this preliminary study, a 3D-printed scaffold conjugated with rhBMP-2 can be used simultaneously as an implant surgical guide and a bone graft in a large bone defect site. Further large-scale studies will be needed to confirm these results.
Collapse
Affiliation(s)
- Ji Cheol Bae
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Jin-Ju Lee
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Jin-Hyung Shim
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-Ro, Siheung 15073, Korea.
| | - Keun-Ho Park
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-Ro, Siheung 15073, Korea.
| | - Jeong-Seok Lee
- Department of Mechanical Engineering, Korea Polytechnic University, 237 Sangidaehak-Ro, Siheung 15073, Korea.
| | - Eun-Bin Bae
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Jae-Won Choi
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| | - Jung-Bo Huh
- Department of Prosthodontics, Dental Research Institute, Institute of Translational Dental Sciences, BK21 PLUS Project, School of Dentistry, Pusan National University, Yangsan 50612, Korea.
| |
Collapse
|
222
|
Lausecker F, Tian X, Inoue K, Wang Z, Pedigo CE, Hassan H, Liu C, Zimmer M, Jinno S, Huckle AL, Hamidi H, Ross RS, Zent R, Ballestrem C, Lennon R, Ishibe S. Vinculin is required to maintain glomerular barrier integrity. Kidney Int 2017; 93:643-655. [PMID: 29241625 PMCID: PMC5846847 DOI: 10.1016/j.kint.2017.09.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 08/23/2017] [Accepted: 09/14/2017] [Indexed: 01/08/2023]
Abstract
Cell-matrix interactions and podocyte intercellular junctions are key for maintaining the glomerular filtration barrier. Vinculin, a cytoplasmic protein, couples actin filaments to integrin-mediated cell-matrix adhesions and to cadherin-based intercellular junctions. Here, we examined the role of vinculin in podocytes by the generation of a podocyte-specific knockout mouse. Mice lacking podocyte vinculin had increased albuminuria and foot process effacement following injury in vivo. Analysis of primary podocytes isolated from the mutant mice revealed defects in cell protrusions, altered focal adhesion size and signaling, as well as impaired cell migration. Furthermore, we found a marked mislocalization of the intercellular junction protein zonula occludens-1. In kidney sections from patients with focal segmental glomerulosclerosis, minimal change disease and membranous nephropathy, we observed dramatic differences in the expression levels and localization of vinculin. Thus, our results suggest that vinculin is necessary to maintain the integrity of the glomerular filtration barrier by modulating podocyte foot processes and stabilizing intercellular junctions.
Collapse
Affiliation(s)
- Franziska Lausecker
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Xuefei Tian
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kazunori Inoue
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zhen Wang
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Christopher E Pedigo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Hossam Hassan
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Chang Liu
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Margaret Zimmer
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Stephanie Jinno
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Abby L Huckle
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hellyeh Hamidi
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Robert S Ross
- Department of Medicine/Cardiology, University of California, San Diego, School of Medicine, La Jolla, California, USA and Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Roy Zent
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Veterans Affairs Hospital, Nashville, Tennessee, USA
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Rachel Lennon
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; School of Biology, Faculty of Biology, Medicine and Health, University of Medicine, Manchester Academic Health Science Centre, UK.
| | - Shuta Ishibe
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
223
|
Liu XQ, Tang RZ. Localized delivery of chemokine for in vitro manipulation of hepatocellular carcinoma cell behaviors during the epithelial–mesenchymal transition. J Biomater Appl 2017; 32:945-956. [DOI: 10.1177/0885328217745774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Xi-Qiu Liu
- Huazhong University of Science and Technology, Tongji Medical College, Wuhan, China
| | - Rui-Zhi Tang
- Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| |
Collapse
|
224
|
Cheng B, Lin M, Huang G, Li Y, Ji B, Genin GM, Deshpande VS, Lu TJ, Xu F. Cellular mechanosensing of the biophysical microenvironment: A review of mathematical models of biophysical regulation of cell responses. Phys Life Rev 2017; 22-23:88-119. [PMID: 28688729 PMCID: PMC5712490 DOI: 10.1016/j.plrev.2017.06.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 06/14/2017] [Indexed: 12/11/2022]
Abstract
Cells in vivo reside within complex microenvironments composed of both biochemical and biophysical cues. The dynamic feedback between cells and their microenvironments hinges upon biophysical cues that regulate critical cellular behaviors. Understanding this regulation from sensing to reaction to feedback is therefore critical, and a large effort is afoot to identify and mathematically model the fundamental mechanobiological mechanisms underlying this regulation. This review provides a critical perspective on recent progress in mathematical models for the responses of cells to the biophysical cues in their microenvironments, including dynamic strain, osmotic shock, fluid shear stress, mechanical force, matrix rigidity, porosity, and matrix shape. The review highlights key successes and failings of existing models, and discusses future opportunities and challenges in the field.
Collapse
Affiliation(s)
- Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Min Lin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Guoyou Huang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Yuhui Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Baohua Ji
- Biomechanics and Biomaterials Laboratory, Department of Applied Mechanics, Beijing Institute of Technology, Beijing, China
| | - Guy M Genin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; Department of Mechanical Engineering & Materials Science, and NSF Science and Technology Center for Engineering Mechanobiology, Washington University in St. Louis, St. Louis 63130, MO, USA
| | - Vikram S Deshpande
- Department of Engineering, University of Cambridge, Cambridge CB2 1PZ, United Kingdom
| | - Tian Jian Lu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
225
|
Panciera T, Azzolin L, Cordenonsi M, Piccolo S. Mechanobiology of YAP and TAZ in physiology and disease. Nat Rev Mol Cell Biol 2017; 18:758-770. [PMID: 28951564 PMCID: PMC6192510 DOI: 10.1038/nrm.2017.87] [Citation(s) in RCA: 924] [Impact Index Per Article: 115.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A growing body of evidence suggests that mechanical signals emanating from the cell's microenvironment are fundamental regulators of cell behaviour. Moreover, at the macroscopic scale, the influence of forces, such as the forces generated by blood flow, muscle contraction, gravity and overall tissue rigidity (for example, inside of a tumour lump), is central to our understanding of physiology and disease pathogenesis. Still, how mechanical cues are sensed and transduced at the molecular level to regulate gene expression has long remained enigmatic. The identification of the transcription factors YAP and TAZ as mechanotransducers started to fill this gap. YAP and TAZ read a broad range of mechanical cues, from shear stress to cell shape and extracellular matrix rigidity, and translate them into cell-specific transcriptional programmes. YAP and TAZ mechanotransduction is critical for driving stem cell behaviour and regeneration, and it sheds new light on the mechanisms by which aberrant cell mechanics is instrumental for the onset of multiple diseases, such as atherosclerosis, fibrosis, pulmonary hypertension, inflammation, muscular dystrophy and cancer.
Collapse
Affiliation(s)
- Tito Panciera
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| | - Luca Azzolin
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| | - Michelangelo Cordenonsi
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua School of Medicine, viale Colombo 3, 35126 Padua, Italy
| |
Collapse
|
226
|
LaValley DJ, Zanotelli MR, Bordeleau F, Wang W, Schwager SC, Reinhart-King CA. Matrix Stiffness Enhances VEGFR-2 Internalization, Signaling, and Proliferation in Endothelial Cells. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017. [PMID: 29531793 DOI: 10.1088/2057-1739/aa9263] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Vascular endothelial growth factor (VEGF) can mediate endothelial cell migration, proliferation, and angiogenesis. During cancer progression, VEGF production is often increased to stimulate the growth of new blood vessels to supply growing tumors with the additional oxygen and nutrients they require. Extracellular matrix stiffening also occurs during tumor progression, however, the crosstalk between tumor mechanics and VEGF signaling remains poorly understood. Here, we show that matrix stiffness heightens downstream endothelial cell response to VEGF by altering VEGF receptor-2 (VEGFR-2) internalization, and this effect is influenced by cell confluency. In sub-confluent endothelial monolayers, VEGFR-2 levels, but not VEGFR-2 phosphorylation, are influenced by matrix rigidity. Interestingly, more compliant matrices correlated with increased expression and clustering of VEGFR-2; however, stiffer matrices induced increased VEGFR-2 internalization. These effects are most likely due to actin-mediated contractility, as inhibiting ROCK on stiff substrates increased VEGFR-2 clustering and decreased internalization. Additionally, increasing matrix stiffness elevates ERK 1/2 phosphorylation, resulting in increased cell proliferation. Moreover, cells on stiff matrices generate more actin stress fibers than on compliant substrates, and the addition of VEGF stimulates an increase in fiber formation regardless of stiffness. In contrast, once endothelial cells reached confluency, stiffness-enhanced VEGF signaling was no longer observed. Together, these data show a complex effect of VEGF and matrix mechanics on VEGF-induced signaling, receptor dynamics, and cell proliferation that is mediated by cell confluency.
Collapse
Affiliation(s)
- Danielle J LaValley
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
| | - Francois Bordeleau
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Cynthia A Reinhart-King
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| |
Collapse
|
227
|
Aghvami M, Billiar KL, Sander EA. Fiber Network Models Predict Enhanced Cell Mechanosensing on Fibrous Gels. J Biomech Eng 2017; 138:2546291. [PMID: 27548709 DOI: 10.1115/1.4034490] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Indexed: 01/07/2023]
Abstract
The propagation of mechanical signals through nonlinear fibrous tissues is much more extensive than through continuous synthetic hydrogels. Results from recent studies indicate that increased mechanical propagation arises from the fibrous nature of the material rather than the strain-stiffening property. The relative importance of different parameters of the fibrous network structure to this propagation, however, remains unclear. In this work, we directly compared the mechanical response of substrates of varying thickness subjected to a constant cell traction force using either a nonfibrous strain-stiffening continuum-based model or a volume-averaged fiber network model consisting of two different types of fiber network structures: one with low fiber connectivity (growth networks) and one with high fiber connectivity (Delaunay networks). The growth network fiber models predicted a greater propagation of substrate displacements through the model and a greater sensitivity to gel thickness compared to the more connected Delaunay networks and the nonlinear continuum model. Detailed analysis of the results indicates that rotational freedom of the fibers in a network with low fiber connectivity is critically important for enhanced, long-range mechanosensing. Our findings demonstrate the utility of multiscale models in predicting cells mechanosensing on fibrous gels, and they provide a more complete understanding of how cell traction forces propagate through fibrous tissues, which has implications for the design of engineered tissues and the stem cell niche.
Collapse
|
228
|
Syed S, Schober J, Blanco A, Zustiak SP. Morphological adaptations in breast cancer cells as a function of prolonged passaging on compliant substrates. PLoS One 2017; 12:e0187853. [PMID: 29136040 PMCID: PMC5685588 DOI: 10.1371/journal.pone.0187853] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 10/29/2017] [Indexed: 11/19/2022] Open
Abstract
Standard tissue culture practices involve propagating cells on tissue culture polystyrene (TCP) dishes, which are flat, 2-dimensional (2D) and orders of magnitude stiffer than most tissues in the body. Such simplified conditions lead to phenotypical cell changes and altered cell behaviors. Hence, much research has been focused on developing novel biomaterials and culture conditions that more closely emulate in vivo cell microenvironments. In particular, biomaterial stiffness has emerged as a key property that greatly affects cell behaviors such as adhesion, morphology, proliferation and motility among others. Here we ask whether cells that have been conditioned to TCP, would still show significant dependence on substrate stiffness if they are first pre-adapted to a more physiologically relevant environment. We used two commonly utilized breast cancer cell lines, namely MDA-MB-231 and MCF-7, and examined the effect of prolonged cell culturing on polyacrylamide substrates of varying compliance. We followed changes in cell adhesion, proliferation, shape factor, spreading area and spreading rate. After pre-adaptation, we noted diminished differences in cell behaviors when comparing between soft (1 kPa) and stiff (103 kPa) gels as well as rigid TCP control. Prolonged culturing of cells on complaint substrates further influenced responses of pre-adapted cells when transferred back to TCP. Our results have implications for the study of stiffness-dependent cell behaviors and indicate that cell pre-adaptation to the substrate needs consideration.
Collapse
Affiliation(s)
- Sana Syed
- Department of Biomedical Engineering, Saint Louis University, Saint Louis, Missouri, United States of America
| | - Joseph Schober
- Department of Pharmaceutical Sciences, Southern Illinois University Edwardsville, Edwardsville, Illinois, United States of America
| | - Alexandra Blanco
- Department of Biomedical Engineering, Saint Louis University, Saint Louis, Missouri, United States of America
| | - Silviya Petrova Zustiak
- Department of Biomedical Engineering, Saint Louis University, Saint Louis, Missouri, United States of America
| |
Collapse
|
229
|
Mazza G, Al-Akkad W, Rombouts K. Engineering in vitro models of hepatofibrogenesis. Adv Drug Deliv Rev 2017; 121:147-157. [PMID: 28578016 DOI: 10.1016/j.addr.2017.05.018] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/17/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
Chronic liver disease is a major cause of morbidity and mortality worldwide marked by chronic inflammation and fibrosis/scarring, resulting in end-stage liver disease and its complications. Hepatic stellate cells (HSCs) are a dominant contributor to liver fibrosis by producing excessive extracellular matrix (ECM), irrespective of the underlying disease aetiologies, and for many decades research has focused on the development of a number of anti-fibrotic strategies targeting this cell. Despite major improvements in two-dimensional systems (2D) by using a variety of cell culture models of different complexity, an efficient anti-fibrogenic therapy has yet to be developed. The development of well-defined three-dimensional (3D) in vitro models, which mimic ECM structures as found in vivo, have demonstrated the importance of cell-matrix bio-mechanics, the complex interactions between HSCs and hepatocytes and other non-parenchymal cells, and this to improve and promote liver cell-specific functions. Henceforth, refinement of these 3D in vitro models, which reproduce the liver microenvironment, will lead to new objectives and to a possible new era in the search for antifibrogenic compounds.
Collapse
|
230
|
Shin YM, Shin HJ, Yang DH, Koh YJ, Shin H, Chun HJ. Advanced capability of radially aligned fibrous scaffolds coated with polydopamine for guiding directional migration of human mesenchymal stem cells. J Mater Chem B 2017; 5:8725-8737. [PMID: 32264266 DOI: 10.1039/c7tb01758h] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In a large tissue defect, faster migration of adjacent tissue toward the defect shortens the tissue regeneration time. Little has been explored on guiding of directional migration from all fronts of the defect boundary towards the center in tissue engineering. This paper demonstrates the effect of radially aligned fibrous scaffolds (RAFSs) coated with polydopamine in order to guide directional migration of human mesenchymal stem cells (hMSCs). RAFSs were electrospun using a collector with a set of electrodes, each constructed with a metallic ring and a point. The polydopamine was then coated by dipping the scaffolds in a dopamine solution (PD-RAFS). The RAFSs exhibited radial distribution of the fibers from the peripheral region toward the center, and polydopamine was uniformly coated over the entire surface by presenting characteristics of the aromatic ring from dopamine. When hMSCs were seeded on the scaffolds, cells grew in an elongated form toward the center along the fiber direction. In particular, the polydopamine coating improved adhesion and spreading of hMSCs on the scaffolds while preserving initial cell orientation. The hMSCs migrated toward the center of the scaffolds at the border of the seeded area; it was enhanced in the order of PD-RAFS > RAFS > random fibrous scaffolds. Therefore, PD-RAFSs can be utilized as an alternate scaffold that can lead to fast and directional migration of cells for finally facilitating tissue regeneration.
Collapse
Affiliation(s)
- Young Min Shin
- Institute of Cell & Tissue Engineering, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| | | | | | | | | | | |
Collapse
|
231
|
Tong Z, Martyn K, Yang A, Yin X, Mead BE, Joshi N, Sherman NE, Langer RS, Karp JM. Towards a defined ECM and small molecule based monolayer culture system for the expansion of mouse and human intestinal stem cells. Biomaterials 2017; 154:60-73. [PMID: 29120819 DOI: 10.1016/j.biomaterials.2017.10.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 10/21/2017] [Accepted: 10/22/2017] [Indexed: 01/04/2023]
Abstract
Current ISC culture systems face significant challenges such as animal-derived or undefined matrix compositions, batch-to-batch variability (e.g. Matrigel-based organoid culture), and complexity of assaying cell aggregates such as organoids which renders the research and clinical translation of ISCs challenging. Here, through screening for suitable ECM components, we report a defined, collagen based monolayer culture system that supports the growth of mouse and human intestinal epithelial cells (IECs) enriched for an Lgr5+ population comparable or higher to the levels found in a standard Matrigel-based organoid culture. The system, referred to as the Bolstering Lgr5 Transformational (BLT) Sandwich culture, comprises a collagen IV-coated porous substrate and a collagen I gel overlay which sandwich an IEC monolayer in between. The distinct collagen cues synergistically regulate IEC attachment, proliferation, and Lgr5 expression through maximizing the engagement of distinct cell surface adhesion receptors (i.e. integrin α2β1, integrin β4) and cell polarity. Further, we apply our BLT Sandwich system to identify that the addition of a bone morphogenetic protein (BMP) receptor inhibitor (LDN-193189) improves the expansion of Lgr5-GFP+ cells from mouse small intestinal crypts by nearly 2.5-fold. Notably, the BLT Sandwich culture is capable of expanding human-derived IECs with higher LGR5 mRNA levels than conventional Matrigel culture, providing superior expansion of human LGR5+ ISCs. Considering the key roles Lgr5+ ISCs play in intestinal epithelial homeostasis and regeneration, we envision that our BLT Sandwich culture system holds great potential for understanding and manipulating ISC biology in vitro (e.g. for modeling ISC-mediated gut diseases) or for expanding a large number of ISCs for clinical utility (e.g. for stem cell therapy).
Collapse
Affiliation(s)
- Zhixiang Tong
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Keir Martyn
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Andy Yang
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Xiaolei Yin
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States; David H. Koch Institute for Integrative Cancer Research at MIT, United States
| | - Benjamin E Mead
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States; Broad Institute of Harvard and MIT, United States; David H. Koch Institute for Integrative Cancer Research at MIT, United States
| | - Nitin Joshi
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Nicholas E Sherman
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States
| | - Robert S Langer
- Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States; Department of Chemical Engineering at MIT, United States; David H. Koch Institute for Integrative Cancer Research at MIT, United States
| | - Jeffrey M Karp
- Division of BioEngineering in Medicine, Department of Medicine, Center for Regenerative Therapeutics, Brigham and Women's Hospital, United States; Harvard Medical School, United States; Harvard Stem Cell Institute, United States; Harvard - Massachusetts Institute of Technology (MIT) Division of Health Sciences and Technology, United States.
| |
Collapse
|
232
|
Cao J, Wang J, Jackman CP, Cox AH, Trembley MA, Balowski JJ, Cox BD, De Simone A, Dickson AL, Di Talia S, Small EM, Kiehart DP, Bursac N, Poss KD. Tension Creates an Endoreplication Wavefront that Leads Regeneration of Epicardial Tissue. Dev Cell 2017; 42:600-615.e4. [PMID: 28950101 DOI: 10.1016/j.devcel.2017.08.024] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 06/24/2017] [Accepted: 08/29/2017] [Indexed: 01/02/2023]
Abstract
Mechanisms that control cell-cycle dynamics during tissue regeneration require elucidation. Here we find in zebrafish that regeneration of the epicardium, the mesothelial covering of the heart, is mediated by two phenotypically distinct epicardial cell subpopulations. These include a front of large, multinucleate leader cells, trailed by follower cells that divide to produce small, mononucleate daughters. By using live imaging of cell-cycle dynamics, we show that leader cells form by spatiotemporally regulated endoreplication, caused primarily by cytokinesis failure. Leader cells display greater velocities and mechanical tension within the epicardial tissue sheet, and experimentally induced tension anisotropy stimulates ectopic endoreplication. Unbalancing epicardial cell-cycle dynamics with chemical modulators indicated autonomous regenerative capacity in both leader and follower cells, with leaders displaying an enhanced capacity for surface coverage. Our findings provide evidence that mechanical tension can regulate cell-cycle dynamics in regenerating tissue, stratifying the source cell features to improve repair.
Collapse
Affiliation(s)
- Jingli Cao
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Jinhu Wang
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Christopher P Jackman
- Regeneration Next, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Amanda H Cox
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Michael A Trembley
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | - Joseph J Balowski
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Ben D Cox
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Alessandro De Simone
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Amy L Dickson
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Stefano Di Talia
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Eric M Small
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14624, USA
| | | | - Nenad Bursac
- Regeneration Next, Duke University, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Kenneth D Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
233
|
Sun Y, Deng R, Zhang K, Ren X, Zhang L, Li J. Single-cell study of the extracellular matrix effect on cell growth by in situ imaging of gene expression. Chem Sci 2017; 8:8019-8024. [PMID: 29568449 PMCID: PMC5855132 DOI: 10.1039/c7sc03880a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 10/01/2017] [Indexed: 12/16/2022] Open
Abstract
The effect of extracellular matrix stiffness on cell growth and the underlying molecular mechanism was investigated using an in situ single-cell imaging of gene expression method based on rolling circle amplification.
Cell behaviors are known to be regulated by the cellular microenvironment. Traditional cell-population based analysis methods need to separate cells from their extracellular matrix (ECM) and cannot resolve the heterogeneity of cell behaviors. Herein, an in situ single-cell analysis method based on rolling circle amplification was exploited to image gene expression in single cells for investigating the effect of ECM stiffness on cell growth. This method enables the simultaneous quantifying of the cell phenotype and gene expression at the single-cell level, which can help in understanding the underlying molecular mechanism of cell growth. It is found that ECM stiffness could affect cell growth via regulating the expression level of the cytoskeleton-assembly associated genes PFN1 and CFL1 and their co-expression pattern. Therefore, this single-cell analysis platform may facilitate us to tap into the study of “single-cell phenotypes” and elucidate the disease association of ECMs.
Collapse
Affiliation(s)
- Yupeng Sun
- Department of Chemistry , Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Ruijie Deng
- Department of Chemistry , Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Kaixiang Zhang
- Department of Chemistry , Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Xiaojun Ren
- Department of Chemistry , Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Ling Zhang
- Department of Chemistry , Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| | - Jinghong Li
- Department of Chemistry , Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology , Tsinghua University , Beijing 100084 , China .
| |
Collapse
|
234
|
Li Y, Tang CB, Kilian KA. Matrix Mechanics Influence Fibroblast-Myofibroblast Transition by Directing the Localization of Histone Deacetylase 4. Cell Mol Bioeng 2017; 10:405-415. [PMID: 31719870 PMCID: PMC6816600 DOI: 10.1007/s12195-017-0493-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 07/07/2017] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION The propagation of mechanochemical signals from the extracellular matrix to the cell nucleus has emerged as a central feature in regulating cellular differentiation and de-differentiation. This process of outside-in signaling and the associated mechanotransduction pathways have been well described in numerous developmental and pathological contexts. However, it remains less clear how mechanotransduction influences the activity of chromatin modifying enzymes that direct gene expression programs. OBJECTIVES The primary objective of this study was to explore how matrix mechanics and geometric confinement influence histone deacetylase (HDAC) activity in fibroblast culture. METHODS Polyacrylamide hydrogels were formed and functionalized with fibronectin patterns using soft lithography. Primary mouse embryonic fibroblasts (MEFs) were cultured on the islands until confluent, fixed, and immunolabeled for microscopy. RESULTS After 24 h MEFs cultured on soft hydrogels (0.5 kPa) show increased expression of class I HDACs relative to MEFs cultured on stiff hydrogels (100 kPa). A member of the class II family, HDAC4 shows a similar trend; however, there is a pronounced cytoplasmic localization on soft hydrogels suggesting a role in outside-in cytoplasmic signaling. Pharmacological inhibitor studies suggest that the opposing activities of extracellular related kinase 1/2 (ERK) and protein phosphatase 2a (PP2a) influence the localization of HDAC4. MEFs cultured on the soft hydrogels show enhanced expression of markers associated with a fibroblast-myofibroblast transition (Paxillin, αSMA). CONCLUSIONS We show that the phosphorylation state and cellular localization of HDAC4 is influenced by matrix mechanics, with evidence for a role in mechanotransduction and the regulation of gene expression associated with fibroblast-myofibroblast transitions. This work establishes a link between outside-in signaling and epigenetic regulation, which will assist efforts aimed at controlling gene regulation in engineered extracellular matrices.
Collapse
Affiliation(s)
- Yanfen Li
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Claire B. Tang
- Department of Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Kristopher A. Kilian
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| |
Collapse
|
235
|
Warren JL, Hoxha E, Jumbo-Lucioni P, De Luca M. Reduction of Syndecan Transcript Levels in the Insulin-Producing Cells Affects Glucose Homeostasis in Adult Drosophila melanogaster. DNA Cell Biol 2017; 36:959-965. [PMID: 28945109 DOI: 10.1089/dna.2017.3912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Signaling by direct cell-matrix interactions has been shown to impact the transcription, secretion, and storage of insulin in mammalian β cells. However, more research is still needed in this area. Syndecans are transmembrane heparan sulfate proteoglycans that function independently and in synergy with integrin-mediated signaling to mediate cell adhesion to the extracellular matrix. In this study, we used the model organism Drosophila melanogaster to determine whether knockdown of the Syndecan (Sdc) gene expression specifically in the insulin-producing cells (IPCs) might affect insulin-like peptide (ILP) production and secretion. IPCs of adult flies produce three ILPs (ILP2, ILP3, and ILP5), which have significant homology to mammalian insulin. We report that flies with reduced Sdc expression in the IPCs did not show any difference in the expression of ilp genes compared to controls. However, they had significantly reduced levels of the circulating ILP2 protein, higher circulating carbohydrates, and were less glucose tolerant than control flies. Finally, we found that IPCs-specific Sdc knockdown led to reduced levels of head Glucose transporter1 gene expression, extracellular signal-regulated kinase phosphorylation, and reactive oxygen species. Taken together, our findings suggest a cell autonomous role for Sdc in insulin release in D. melanogaster.
Collapse
Affiliation(s)
- Jonathan L Warren
- 1 Department of Nutrition Sciences, University of Alabama at Birmingham , Birmingham, Alabama
| | - Eneida Hoxha
- 1 Department of Nutrition Sciences, University of Alabama at Birmingham , Birmingham, Alabama.,2 Department of Biology, Ecology and Earth Science, University of Calabria , Rende, Italy
| | - Patricia Jumbo-Lucioni
- 1 Department of Nutrition Sciences, University of Alabama at Birmingham , Birmingham, Alabama.,3 Department of Pharmaceutical, Social and Administrative Sciences, McWhorter School of Pharmacy, Samford University , Birmingham, Alabama
| | - Maria De Luca
- 1 Department of Nutrition Sciences, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
236
|
Low level of ochratoxin A affects genome-wide expression in kidney of pig. Toxicon 2017; 136:67-77. [PMID: 28688805 DOI: 10.1016/j.toxicon.2017.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/30/2017] [Accepted: 07/04/2017] [Indexed: 01/18/2023]
|
237
|
McKee C, Chaudhry GR. Advances and challenges in stem cell culture. Colloids Surf B Biointerfaces 2017; 159:62-77. [PMID: 28780462 DOI: 10.1016/j.colsurfb.2017.07.051] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 07/04/2017] [Accepted: 07/22/2017] [Indexed: 12/12/2022]
Abstract
Stem cells (SCs) hold great promise for cell therapy, tissue engineering, and regenerative medicine as well as pharmaceutical and biotechnological applications. They have the capacity to self-renew and the ability to differentiate into specialized cell types depending upon their source of isolation. However, use of SCs for clinical applications requires a high quality and quantity of cells. This necessitates large-scale expansion of SCs followed by efficient and homogeneous differentiation into functional derivatives. Traditional methods for maintenance and expansion of cells rely on two-dimensional (2-D) culturing techniques using plastic culture plates and xenogenic media. These methods provide limited expansion and cells tend to lose clonal and differentiation capacity upon long-term passaging. Recently, new approaches for the expansion of SCs have emphasized three-dimensional (3-D) cell growth to mimic the in vivo environment. This review provides a comprehensive compendium of recent advancements in culturing SCs using 2-D and 3-D techniques involving spheroids, biomaterials, and bioreactors. In addition, potential challenges to achieve billion-fold expansion of cells are discussed.
Collapse
Affiliation(s)
- Christina McKee
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA
| | - G Rasul Chaudhry
- Department of Biological Sciences , Oakland University, Rochester, MI, 48309, USA; OU-WB Institute for Stem Cell and Regenerative Medicine, Oakland University, Rochester, MI, 48309, USA.
| |
Collapse
|
238
|
Golfier S, Rosendahl P, Mietke A, Herbig M, Guck J, Otto O. High-throughput cell mechanical phenotyping for label-free titration assays of cytoskeletal modifications. Cytoskeleton (Hoboken) 2017; 74:283-296. [PMID: 28445605 PMCID: PMC5601209 DOI: 10.1002/cm.21369] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/12/2017] [Accepted: 04/20/2017] [Indexed: 01/29/2023]
Abstract
The mechanical fingerprint of cells is inherently linked to the structure of the cytoskeleton and can serve as a label‐free marker for cell homeostasis or pathologic states. How cytoskeletal composition affects the physical response of cells to external loads has been intensively studied with a spectrum of techniques, yet quantitative and statistically powerful investigations in the form of titration assays are hampered by the low throughput of most available methods. In this study, we employ real‐time deformability cytometry (RT‐DC), a novel microfluidic tool to examine the effects of biochemically modified F‐actin and microtubule stability and nuclear chromatin structure on cell deformation in a human leukemia cell line (HL60). The high throughput of our method facilitates extensive titration assays that allow for significance assessment of the observed effects and extraction of half‐maximal concentrations for most of the applied reagents. We quantitatively show that integrity of the F‐actin cortex and microtubule network dominate cell deformation on millisecond timescales probed with RT‐DC. Drug‐induced alterations in the nuclear chromatin structure were not found to consistently affect cell deformation. The sensitivity of the high‐throughput cell mechanical measurements to the cytoskeletal modifications we present in this study opens up new possibilities for label‐free dose‐response assays of cytoskeletal modifications.
Collapse
Affiliation(s)
- Stefan Golfier
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany.,Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Max-Planck-Institute for Physics of Complex Systems, Dresden, Germany
| | - Philipp Rosendahl
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Alexander Mietke
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Max-Planck-Institute for Physics of Complex Systems, Dresden, Germany
| | - Maik Herbig
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Jochen Guck
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Oliver Otto
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany.,ZIK HIKE, Universität Greifswald, Greifswald, Germany
| |
Collapse
|
239
|
Beamish JA, Chen E, Putnam AJ. Engineered extracellular matrices with controlled mechanics modulate renal proximal tubular cell epithelialization. PLoS One 2017; 12:e0181085. [PMID: 28715434 PMCID: PMC5513452 DOI: 10.1371/journal.pone.0181085] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 06/26/2017] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is common and associated with significant morbidity and mortality. Recovery from many forms of AKI involves the proliferation of renal proximal tubular epithelial cells (RPTECs), but the influence of the microenvironment in which this recovery occurs remains poorly understood. Here we report the development of a poly(ethylene glycol) (PEG) hydrogel platform to study the influence of substrate mechanical properties on the proliferation of human RPTECs as a model for recovery from AKI. PEG diacrylate based hydrogels were generated with orthogonal control of mechanics and cell-substrate interactions. Using this platform, we found that increased substrate stiffness promotes RPTEC spreading and proliferation. RPTECs showed similar degrees of apoptosis and Yes-associated protein (YAP) nuclear localization regardless of stiffness, suggesting these were not key mediators of the effect. However, focal adhesion formation, cytoskeletal organization, focal adhesion kinase (FAK) activation, and extracellular signal-regulated kinase (ERK) activation were all enhanced with increasing substrate stiffness. Inhibition of ERK activation substantially attenuated the effect of stiffness on proliferation. In long-term culture, hydrogel stiffness promoted the formation of more complete epithelial monolayers with tight junctions, cell polarity, and an organized basement membrane. These data suggest that increased stiffness potentially may have beneficial consequences for the renal tubular epithelium during recovery from AKI.
Collapse
Affiliation(s)
- Jeffrey A. Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Evan Chen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
240
|
Zhou Z, Sun X, Ma J, Tong M, To S, Wong A, Ngan A. Actin cytoskeleton stiffness grades metastatic potential of ovarian carcinoma Hey A8 cells via nanoindentation mapping. J Biomech 2017; 60:219-226. [DOI: 10.1016/j.jbiomech.2017.06.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/24/2017] [Accepted: 06/25/2017] [Indexed: 02/03/2023]
|
241
|
Heo SJ, Han WM, Szczesny SE, Cosgrove BD, Elliott DM, Lee DA, Duncan RL, Mauck RL. Mechanically Induced Chromatin Condensation Requires Cellular Contractility in Mesenchymal Stem Cells. Biophys J 2017; 111:864-874. [PMID: 27558729 DOI: 10.1016/j.bpj.2016.07.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/27/2016] [Accepted: 07/11/2016] [Indexed: 02/07/2023] Open
Abstract
Mechanical cues play important roles in directing the lineage commitment of mesenchymal stem cells (MSCs). In this study, we explored the molecular mechanisms by which dynamic tensile loading (DL) regulates chromatin organization in this cell type. Our previous findings indicated that the application of DL elicited a rapid increase in chromatin condensation through purinergic signaling mediated by ATP. Here, we show that the rate and degree of condensation depends on the frequency and duration of mechanical loading, and that ATP release requires actomyosin-based cellular contractility. Increases in baseline cellular contractility via the addition of an activator of G-protein coupled receptors (lysophosphatidic acid) induced rapid ATP release, resulting in chromatin condensation independent of loading. Conversely, inhibition of contractility through pretreatment with either a RhoA/Rock inhibitor (Y27632) or MLCK inhibitor (ML7) abrogated ATP release in response to DL, blocking load-induced chromatin condensation. With loading, ATP release occurred very rapidly (within the first 10-20 s), whereas changes in chromatin occurred at a later time point (∼10 min), suggesting a downstream biochemical pathway mediating this process. When cells were pretreated with blockers of the transforming growth factor (TGF) superfamily, purinergic signaling in response to DL was also eliminated. Further analysis showed that this pretreatment decreased contractility, implicating activity in the TGF pathway in the establishment of the baseline contractile state of MSCs (in the absence of exogenous ligands). These data indicate that chromatin condensation in response to DL is regulated through the interplay between purinergic and RhoA/Rock signaling, and that ligandless activity in the TGF/bone morphogenetic proteins signaling pathway contributes to the establishment of baseline contractility in MSCs.
Collapse
Affiliation(s)
- Su-Jin Heo
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Woojin M Han
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Spencer E Szczesny
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, Pennsylvania
| | - Brian D Cosgrove
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, Pennsylvania
| | - Dawn M Elliott
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware
| | - David A Lee
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, United Kingdom
| | - Randall L Duncan
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware; Department of Biological Sciences, University of Delaware, Newark, Delaware
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania; Translational Musculoskeletal Research Center, Philadelphia VA Medical Center, Philadelphia, Pennsylvania.
| |
Collapse
|
242
|
Iwata N, Nozaki K, Horiuchi N, Yamashita K, Tsutsumi Y, Miura H, Nagai A. Effects of controlled micro-/nanosurfaces on osteoblast proliferation. J Biomed Mater Res A 2017; 105:2589-2596. [DOI: 10.1002/jbm.a.36118] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Natsuko Iwata
- Department of Fixed Prosthodontics; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University; Tokyo Japan
| | - Kosuke Nozaki
- Department of Biofunction Research; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University; Tokyo Japan
| | - Naohiro Horiuchi
- Department of Inorganic Biomaterials; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University; Tokyo Japan
| | - Kimihiro Yamashita
- Department of Inorganic Biomaterials; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University; Tokyo Japan
| | - Yusuke Tsutsumi
- Department of Metallic Biomaterials; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University; Tokyo Japan
| | - Hiroyuki Miura
- Department of Fixed Prosthodontics; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University; Tokyo Japan
| | - Akiko Nagai
- Department of Biofunction Research; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University; Tokyo Japan
| |
Collapse
|
243
|
Wang X, Lee J, Ali M, Kim J, Lacerda CMR. Phenotype Transformation of Aortic Valve Interstitial Cells Due to Applied Shear Stresses Within a Microfluidic Chip. Ann Biomed Eng 2017. [DOI: 10.1007/s10439-017-1871-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
244
|
Bell S, Terentjev EM. Focal Adhesion Kinase: The Reversible Molecular Mechanosensor. Biophys J 2017; 112:2439-2450. [PMID: 28591616 PMCID: PMC5474844 DOI: 10.1016/j.bpj.2017.04.048] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/31/2017] [Accepted: 04/28/2017] [Indexed: 12/19/2022] Open
Abstract
Sensors are the first element of the pathways that control the response of cells to their environment. Protein complexes that produce or enable a chemical signal in response to a mechanical stimulus are called "mechanosensors". In this work, we develop a theoretical model describing the physical mechanism of a reversible single-molecule stiffness sensor. Although this has the potential for general application, here we apply the model to focal adhesion kinase, which initiates the chemical signal in its active phosphorylated conformation, but can spontaneously return to its closed folded conformation. We find how the rates of conformation changes depend on the substrate stiffness and the pulling force applied from the cell cytoskeleton. We find the sensor is homeostatic, spontaneously self-adjusting to reach a state where its range of maximum sensitivity matches the substrate stiffness. The results compare well with the phenotype observations of cells on different substrates.
Collapse
Affiliation(s)
- Samuel Bell
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Eugene M Terentjev
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
245
|
Provenzano PP. Tug of War at the Cell-Matrix Interface. Biophys J 2017; 112:1739-1741. [PMID: 28494945 DOI: 10.1016/j.bpj.2017.03.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 03/31/2017] [Indexed: 10/19/2022] Open
Affiliation(s)
- Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota; Physical Sciences in Oncology Center, University of Minnesota, Minneapolis, Minnesota; Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota; Institute for Engineering in Medicine, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
246
|
Zhou DW, Lee TT, Weng S, Fu J, García AJ. Effects of substrate stiffness and actomyosin contractility on coupling between force transmission and vinculin-paxillin recruitment at single focal adhesions. Mol Biol Cell 2017; 28:1901-1911. [PMID: 28468976 PMCID: PMC5541841 DOI: 10.1091/mbc.e17-02-0116] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/31/2017] [Accepted: 04/27/2017] [Indexed: 12/22/2022] Open
Abstract
The relationship between force and focal adhesion (FA) dynamics is unclear. Substrate stiffness and contractility regulate the relationship between force and vinculin, but not paxillin, turnover at FAs. Substrate stiffness and contractility also regulate whether vinculin and paxillin turnover dynamics are correlated at FAs. Focal adhesions (FAs) regulate force transfer between the cytoskeleton and ECM–integrin complexes. We previously showed that vinculin regulates force transmission at FAs. Vinculin residence time in FAs correlated with applied force, supporting a mechanosensitive model in which forces stabilize vinculin’s active conformation to promote force transfer. In the present study, we examined the relationship between traction force and vinculin–paxillin localization to single FAs in the context of substrate stiffness and actomyosin contractility. We found that vinculin and paxillin FA area did not correlate with traction force magnitudes at single FAs, and this was consistent across different ECM stiffness and cytoskeletal tension states. However, vinculin residence time at FAs varied linearly with applied force for stiff substrates, and this was disrupted on soft substrates and after contractility inhibition. In contrast, paxillin residence time at FAs was independent of local applied force and substrate stiffness. Paxillin recruitment and residence time at FAs, however, were dependent on cytoskeletal contractility on lower substrate stiffness values. Finally, substrate stiffness and cytoskeletal contractility regulated whether vinculin and paxillin turnover dynamics are correlated to each other at single FAs. This analysis sheds new insights on the coupling among force, substrate stiffness, and FA dynamics.
Collapse
Affiliation(s)
- Dennis W Zhou
- Wallace H. Coulter Department of Biomedical Engineering, Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA 30332
| | - Ted T Lee
- Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| | - Shinuo Weng
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Jianping Fu
- Department of Biomedical Engineering and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109.,Department of Biomedical Engineering and Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332
| |
Collapse
|
247
|
Abbas Y, Oefner CM, Polacheck WJ, Gardner L, Farrell L, Sharkey A, Kamm R, Moffett A, Oyen ML. A microfluidics assay to study invasion of human placental trophoblast cells. J R Soc Interface 2017; 14:20170131. [PMID: 28566515 PMCID: PMC5454302 DOI: 10.1098/rsif.2017.0131] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/04/2017] [Indexed: 12/22/2022] Open
Abstract
Pre-eclampsia, fetal growth restriction and stillbirth are major pregnancy disorders throughout the world. The underlying pathogenesis of these diseases is defective placentation characterized by inadequate invasion of extravillous placental trophoblast cells into the uterine arteries. How trophoblast invasion is controlled remains an unanswered question but is influenced by maternal uterine immune cells called decidual natural killer cells. Here, we describe an in vitro microfluidic invasion assay to study the migration of primary human trophoblast cells. Each experiment can be performed with a small number of cells making it possible to conduct research on human samples despite the challenges of isolating primary trophoblast cells. Cells are exposed to a chemical gradient and tracked in a three-dimensional microenvironment using real-time high-resolution imaging, so that dynamic readouts on cell migration such as directionality, motility and velocity are obtained. The microfluidic system was validated using isolated trophoblast and a gradient of granulocyte-macrophage colony-stimulating factor, a cytokine produced by activated decidual natural killer cells. This microfluidic model provides detailed analysis of the dynamics of trophoblast migration compared to previous assays and can be modified in future to study in vitro how human trophoblast behaves during placentation.
Collapse
Affiliation(s)
- Yassen Abbas
- The Nanoscience Centre, Department of Engineering, University of Cambridge, Cambridge CB3 0FF, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Carolin Melati Oefner
- The Nanoscience Centre, Department of Engineering, University of Cambridge, Cambridge CB3 0FF, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | | | - Lucy Gardner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Lydia Farrell
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Andrew Sharkey
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Roger Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Michelle L Oyen
- The Nanoscience Centre, Department of Engineering, University of Cambridge, Cambridge CB3 0FF, UK
- Centre for Trophoblast Research (CTR), Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
248
|
Liu XQ, Fourel L, Dalonneau F, Sadir R, Leal S, Lortat-Jacob H, Weidenhaupt M, Albiges-Rizo C, Picart C. Biomaterial-enabled delivery of SDF-1α at the ventral side of breast cancer cells reveals a crosstalk between cell receptors to promote the invasive phenotype. Biomaterials 2017; 127:61-74. [PMID: 28279922 PMCID: PMC5777630 DOI: 10.1016/j.biomaterials.2017.02.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 02/17/2017] [Accepted: 02/26/2017] [Indexed: 12/31/2022]
Abstract
The SDF-1α chemokine (CXCL12) is a potent bioactive chemoattractant known to be involved in hematopoietic stem cell homing and cancer progression. The associated SDF-1α/CXCR4 receptor signaling is a hallmark of aggressive tumors, which can metastasize to distant sites such as lymph nodes, lung and bone. Here, we engineered a biomimetic tumoral niche made of a thin and soft polyelectrolyte film that can retain SDF-1α to present it, in a spatially-controlled manner, at the ventral side of the breast cancer cells. Matrix-bound SDF-1α but not soluble SDF-1α induced a striking increase in cell spreading and migration in a serum-containing medium, which was associated with the formation of lamellipodia and filopodia in MDA-MB231 cells and specifically mediated by CXCR4. Other Knockdown and inhibition experiments revealed that CD44, the major hyaluronan receptor, acted in concert, via a spatial coincidence, to drive a specific matrix-bound SDFα-induced cell response associated with ERK signaling. In contrast, the β1 integrin adhesion receptor played only a minor role on cell polarity. The CXCR4/CD44 mediated cellular response to matrix-bound SDF-1α involved the Rac1 RhoGTPase and was sustained solely in the presence of matrix-bound SDFα, in contrast with the transient signaling observed in response to soluble SDF-1α. Our results highlight that a biomimetic tumoral niche enables to reveal potent cellular effects and so far hidden molecular mechanisms underlying the breast cancer response to chemokines. These results open new insights for the design of future innovative therapies in metastatic cancers, by inhibiting CXCR4-mediated signaling in the tumoral niche via dual targeting of receptors (CXCR4 and CD44) or of associated signaling molecules (CXCR4 and Rac1).
Collapse
Affiliation(s)
- Xi Qiu Liu
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France; FONDATION ARC, 9 rue Guy Môquet, 94803, Villejuif, France; Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Laure Fourel
- Inserm U1209, Université Grenoble Alpes, Institut Albert Bonniot, Site Santé, 38042, Grenoble cedex 9, France; CNRS UMR5309, Institute for Advanced Biosciences, Institut Albert Bonniot, 38700, La Tronche, France
| | - Fabien Dalonneau
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Rabia Sadir
- Institut de Biologie Structurale, UMR 5075, Univ. Grenoble Alpes, CNRS, CEA, F-38027, Grenoble, France
| | - Salome Leal
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Hugues Lortat-Jacob
- Institut de Biologie Structurale, UMR 5075, Univ. Grenoble Alpes, CNRS, CEA, F-38027, Grenoble, France
| | - Marianne Weidenhaupt
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France
| | - Corinne Albiges-Rizo
- Inserm U1209, Université Grenoble Alpes, Institut Albert Bonniot, Site Santé, 38042, Grenoble cedex 9, France; CNRS UMR5309, Institute for Advanced Biosciences, Institut Albert Bonniot, 38700, La Tronche, France
| | - Catherine Picart
- CNRS UMR 5628 (LMGP), 3 parvis Louis Néel, 38016, Grenoble, France; Université Grenoble Alpes, LMGP, 3 parvis Louis Néel, 38016, Grenoble, France.
| |
Collapse
|
249
|
Actomyosin contractility provokes contact inhibition in E-cadherin-ligated keratinocytes. Sci Rep 2017; 7:46326. [PMID: 28406163 PMCID: PMC5390311 DOI: 10.1038/srep46326] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 03/15/2017] [Indexed: 01/09/2023] Open
Abstract
Confluence-dependent inhibition of epithelial cell proliferation, termed contact inhibition, is crucial for epithelial homeostasis and organ size control. Here we report that among epithelial cells, keratinocytes, which compose the stratified epithelium in the skin, possess a unique, actomyosin-dependent mechanism for contact inhibition. We have observed that under actomyosin-inhibited conditions, cell-cell contact itself through E-cadherin promotes proliferation of keratinocytes. Actomyosin activity in confluent keratinocytes, however, inhibits nuclear localization of β-catenin and YAP, and causes attenuation of β-catenin- and YAP-driven cell proliferation. Confluent keratinocytes develop E-cadherin-mediated punctate adhesion complexes, to which radial actin cables are connected. Eliminating the actin-to-E-cadherin linkage by depleting α-catenin increases proliferation of confluent keratinocytes. By contrast, enforced activation of RhoA-regulated actomyosin or external application of pulling force to ligated E-cadherin attenuates their proliferation, suggesting that tensile stress at E-cadherin-mediated adhesion complexes inhibits proliferation of confluent keratinocytes. Our results highlight actomyosin contractility as a crucial factor that provokes confluence-dependent inhibition of keratinocyte proliferation.
Collapse
|
250
|
Virjula S, Zhao F, Leivo J, Vanhatupa S, Kreutzer J, Vaughan TJ, Honkala AM, Viehrig M, Mullen CA, Kallio P, McNamara LM, Miettinen S. The effect of equiaxial stretching on the osteogenic differentiation and mechanical properties of human adipose stem cells. J Mech Behav Biomed Mater 2017; 72:38-48. [PMID: 28448920 DOI: 10.1016/j.jmbbm.2017.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/05/2017] [Accepted: 04/12/2017] [Indexed: 01/22/2023]
Abstract
Although mechanical cues are known to affect stem cell fate and mechanobiology, the significance of such stimuli on the osteogenic differentiation of human adipose stem cells (hASCs) remains unclear. In this study, we investigated the effect of long-term mechanical stimulation on the attachment, osteogenic differentiation and mechanical properties of hASCs. Tailor-made, pneumatic cell stretching devices were used to expose hASCs to cyclic equiaxial stretching in osteogenic medium. Cell attachment and focal adhesions were visualised using immunocytochemical vinculin staining on days 3 and 6, and the proliferation and alkaline phosphatase activity, as a sign of early osteogenic differentiation, were analysed on days 0, 6 and 10. Furthermore, the mechanical properties of hASCs, in terms of apparent Young's modulus and normalised contractility, were obtained using a combination of atomic force microscopy based indentation and computational approaches. Our results indicated that cyclic equiaxial stretching delayed proliferation and promoted osteogenic differentiation of hASCs. Stretching also reduced cell size and intensified focal adhesions and actin cytoskeleton. Moreover, cell stiffening was observed during osteogenic differentiation and especially under mechanical stimulation. These results suggest that cyclic equiaxial stretching modifies cell morphology, focal adhesion formation and mechanical properties of hASCs. This could be exploited to enhance osteogenic differentiation.
Collapse
Affiliation(s)
- Sanni Virjula
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland; Science Centre, Tampere University Hospital, Biokatu 6, 33520 Tampere, Finland.
| | - Feihu Zhao
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| | - Joni Leivo
- Department of Automation Science and Engineering, BioMediTech, Tampere University of Technology, Korkeakoulunkatu 3, 33720 Tampere, Finland.
| | - Sari Vanhatupa
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland; Science Centre, Tampere University Hospital, Biokatu 6, 33520 Tampere, Finland.
| | - Joose Kreutzer
- Department of Automation Science and Engineering, BioMediTech, Tampere University of Technology, Korkeakoulunkatu 3, 33720 Tampere, Finland.
| | - Ted J Vaughan
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| | - Anna-Maija Honkala
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland; Science Centre, Tampere University Hospital, Biokatu 6, 33520 Tampere, Finland.
| | - Marlitt Viehrig
- Department of Automation Science and Engineering, BioMediTech, Tampere University of Technology, Korkeakoulunkatu 3, 33720 Tampere, Finland.
| | - Conleth A Mullen
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| | - Pasi Kallio
- Department of Automation Science and Engineering, BioMediTech, Tampere University of Technology, Korkeakoulunkatu 3, 33720 Tampere, Finland.
| | - Laoise M McNamara
- Biomechanics Research Centre (BMEC), Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland.
| | - Susanna Miettinen
- Adult Stem Cell Group, BioMediTech, Faculty of Medicine and Life Sciences, University of Tampere, Lääkärinkatu 1, 33520 Tampere, Finland; Science Centre, Tampere University Hospital, Biokatu 6, 33520 Tampere, Finland.
| |
Collapse
|