1
|
Dutta Gupta S, Pal N, Ta M. Vitronectin regulates focal adhesion turnover and migration of human placenta-derived MSCs under nutrient stress. Eur J Cell Biol 2025; 104:151477. [PMID: 39893799 DOI: 10.1016/j.ejcb.2025.151477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/07/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
At sites of tissue damage and wound healing, the mesenchymal stem cells (MSCs) are often challenged by nutrient availability due to blood supply disruption. Thus, it becomes critical to identify novel factors and their mechanism of action in regulating the adhesion and migration of MSCs under nutrient stress condition for successful clinical application. In human placenta-derived MSCs (PL-MSCs), we demonstrated an increase in cell spread area, along with increased adhesion and reduced migration of the cells, when cultured under nutrient stress condition. Correspondingly, an increase in the total number per cell and size of focal adhesions (FAs), together with prominent stress fibers were observed in nutrient-stressed PL-MSCs compared to control PL-MSCs. The FAs were demonstrated to be more stable, exhibiting slower turnover and longer lifespan. Vitronectin (VTN), an ECM glycoprotein, was upregulated under nutrient stress condition. Knockdown of VTN in PL-MSCs led to a significant reduction in the total number per cell and size of FAs, along with their faster turnover and shorter lifespan. Subsequently, a reversal in the cell spread area, adhesion and migration properties of the nutrient-stressed PL-MSCs were noted. Additionally, our findings indicated that VTN, as an upstream regulator, stimulated the phosphorylation of myosin light chain, which possibly promoted the maturation and stability of FAs along with assembly of stress fibers, thereby leading to increased adhesion and reduced migration of the cells. Overall, our study defines a distinct role of VTN as a critical regulator of migration in PL-MSCs under nutrient stress condition.
Collapse
Affiliation(s)
- Srishti Dutta Gupta
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| | - Nitish Pal
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| | - Malancha Ta
- Indian Institute of Science Education and Research, Kolkata (IISER Kolkata), India.
| |
Collapse
|
2
|
Saporito S, Panzetta V, Netti PA. Time and space modulation of substrate curvature to regulate cell mechanical identity. Acta Biomater 2024; 186:300-315. [PMID: 39127326 DOI: 10.1016/j.actbio.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/27/2024] [Revised: 07/08/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Recently, a variety of microenvironmental biophysical stimuli have been proved to play a crucial role in regulating cell functions. Among them, morpho-physical cues, like curvature, are emerging as key regulators of cellular behavior. Changes in substrate curvature have been shown to impact the arrangement of Focal Adhesions (FAs), influencing the direction and intensity of cytoskeleton generated forces and resulting in an overall alteration of cell mechanical identity. In their native environment, cells encounter varying degrees of substrate curvature, and in specific organs, they are exposed to dynamic changes of curvature due to periodic tissue deformation. However, the mechanism by which cells perceive substrate curvature remains poorly understood. To this aim, a micro-pneumatic device was designed and implemented. This device enables the controlled application of substrate curvature, both statically and dynamically. Employing a combined experimental and simulative approach, human adipose-derived stem cells were exposed to controlled curvature intensity and frequency. During this exposure, measurements were taken on FAs extension and orientation, cytoskeleton organization and cellular/nuclear alignment. The data clearly indicated a significant influence of the substrate curvature on cell adhesion processes. These findings contribute to a better understanding of the mechanisms through which cells perceive and respond to substrate curvature signals. STATEMENT OF SIGNIFICANCE: This work is our contribution to the comprehension of substrate curvature's function as a crucial regulator of cell adhesion at the scale of focal adhesions and cell mechanical identity. In recent years, a large body of knowledge is continuously growing providing comprehension of the role of various microenvironmental biophysical stimuli in regulating cell functions. Nevertheless, little is known about the role of substrate curvature, in particular, when cells are exposed to this stimulus in a dynamic manner. To address the role of substrate curvature on cellular behavior, a micro-pneumatic device was designed and implemented. This device enables the controlled application of substrate curvature, both statically and dynamically. The experiment data made it abundantly evident that the substrate curvature had a major impact on the mechanisms involved in cell adhesion.
Collapse
Affiliation(s)
- Stefania Saporito
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy
| | - Valeria Panzetta
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy; Interdisciplinary research Center on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy; Interdisciplinary research Center on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy.
| |
Collapse
|
3
|
Xue R, Chen Y, Gong Z, Jiang H. Superposition of Substrate Deformation Fields Induced by Molecular Clutches Explains Cell Spatial Sensing of Ligands. ACS NANO 2024. [PMID: 39088555 DOI: 10.1021/acsnano.4c03667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 08/03/2024]
Abstract
Cells can sense the physical properties of the extracellular matrices (ECMs), such as stiffness and ligand density, through cell adhesions to actively regulate their behaviors. Recent studies have shown that varying ligand spacing of ECMs can influence adhesion size, cell spreading, and even stem cell differentiation, indicating that cells have the spatial sensing ability of ECM ligands. However, the mechanism of the cells' spatial sensing remains unclear. In this study, we have developed a lattice-spring motor-clutch model by integrating cell membrane deformation, the talin unfolding mechanism, and the lattice spring for substrate ligand distribution to explore how the spatial distribution of integrin ligands and substrate stiffness influence cell spreading and adhesion dynamics. By applying the Gillespie algorithm, we found that large ligand spacing reduces the superposition effect of the substrate's displacement fields generated by pulling force from motor-clutch units, increasing the effective stiffness probed by the force-sensitive receptors; this finding explains a series of previous experiments. Furthermore, using the mean-field theory, we obtain the effective stiffness sensed by bound clutches analytically; our analysis shows that the bound clutch number and ligand spacing are the two key factors that affect the superposition effects of deformation fields and, hence, the effective stiffness. Overall, our study reveals the mechanism of cells' spatial sensing, i.e., ligand spacing changes the effective stiffness sensed by cells due to the superposition effect of deformation fields, which provides a physical clue for designing and developing biological materials that effectively control cell behavior and function.
Collapse
Affiliation(s)
- Ruihao Xue
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Yonggang Chen
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Ze Gong
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, China
- State Key Laboratory of Nonlinear Mechanics, Institute of Mechanics, Chinese Academy of Sciences, 15 Beisihuan West Road, Beijing 100190, China
| | - Hongyuan Jiang
- CAS Key Laboratory of Mechanical Behavior and Design of Materials, Department of Modern Mechanics, University of Science and Technology of China, Hefei, Anhui 230027, China
| |
Collapse
|
4
|
Lammert FC, Pannhausen J, Noetzel E, Friedland F, Wirtz J, Herfs Y, Leypold S, Gan L, Weiskirchen R, Schnitzler T, Knüchel R, Maurer J, Jonigk DD, Rose M, Gaisa NT. Dual role of GRHL3 in bladder carcinogenesis depending on histological subtypes. Mol Oncol 2024; 18:1397-1416. [PMID: 38429970 PMCID: PMC11164254 DOI: 10.1002/1878-0261.13623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/15/2023] [Revised: 01/23/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024] Open
Abstract
The effect of grainyhead-like transcription factor 3 (GRHL3) on cancer development depends on the cancer subtypes as shown in tumor entities such as colorectal or oral squamous cell carcinomas. Here, we analyzed the subtype-specific role of GRHL3 in bladder carcinogenesis, comparing common urothelial carcinoma (UC) with squamous bladder cancer (sq-BLCA). We examined GRHL3 mRNA and protein expression in cohorts of patient samples, its prognostic role and its functional impact on tumorigeneses in different molecular and histopathological subtypes of bladder cancer. We showed for GRHL3 a reverse expression in squamous and urothelial bladder cancer subtypes. Stably GRHL3-overexpressing EJ28, J82, and SCaBER in vitro models revealed a tumor-suppressive function in squamous and an oncogenic role in the urothelial cancer cells affecting cell and colony growth, and migratory and invasive capacities. Transcriptomic profiling demonstrated highly subtype-specific GRHL3-regulated expression networks coined by the enrichment of genes involved in integrin-mediated pathways. In SCaBER, loss of ras homolog family member A (RHOA) GTPase activity was demonstrated to be associated with co-regulation of eukaryotic translation initiation factor 4E family member 3 (EIF4E3), a potential tumor suppressor gene. Thus, our data provide for the first time a detailed insight into the role of the transcription factor GRHL3 in different histopathological subtypes of bladder cancer.
Collapse
Affiliation(s)
- Franziska C. Lammert
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Julia Pannhausen
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Erik Noetzel
- Institute of Biological Information Processing 2 (IBI‐2), Mechanobiology, Forschungszentrum Jülich GmbHGermany
| | - Florian Friedland
- Institute of Biological Information Processing 2 (IBI‐2), Mechanobiology, Forschungszentrum Jülich GmbHGermany
| | - Julia Wirtz
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Yannick Herfs
- Institute of Biological Information Processing 2 (IBI‐2), Mechanobiology, Forschungszentrum Jülich GmbHGermany
| | - Sophie Leypold
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
| | - Lin Gan
- IZKF AachenMedical Faculty of the RWTH Aachen UniversityGermany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), University HospitalRWTH Aachen UniversityGermany
| | - Tician Schnitzler
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
| | - Ruth Knüchel
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
| | - Jochen Maurer
- Department of Obstetrics and GynecologyUniversity Hospital AachenGermany
| | - Danny D. Jonigk
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- German Center for Lung Research, DZL, BREATHHanoverGermany
| | - Michael Rose
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- Institute of Pathology, University HospitalUniversity of UlmGermany
| | - Nadine T. Gaisa
- Institute of Pathology, University HospitalRWTH Aachen UniversityGermany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD)Germany
- Institute of Pathology, University HospitalUniversity of UlmGermany
| |
Collapse
|
5
|
Djakbarova U, Madraki Y, Chan ET, Wu T, Atreaga-Muniz V, Akatay AA, Kural C. Tension-induced adhesion mode switching: the interplay between focal adhesions and clathrin-containing adhesion complexes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.07.579324. [PMID: 38370749 PMCID: PMC10871318 DOI: 10.1101/2024.02.07.579324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 02/20/2024]
Abstract
Integrin-based adhesion complexes are crucial in various cellular processes, including proliferation, differentiation, and motility. While the dynamics of canonical focal adhesion complexes (FAs) have been extensively studied, the regulation and physiological implications of the recently identified clathrin-containing adhesion complexes (CCACs) are still not well understood. In this study, we investigated the spatiotemporal mechanoregulations of FAs and CCACs in a breast cancer model. Employing single-molecule force spectroscopy coupled with live-cell fluorescence microscopy, we discovered that FAs and CCACs are mutually exclusive and inversely regulated complexes. This regulation is orchestrated through the modulation of plasma membrane tension, in combination with distinct modes of actomyosin contractility that can either synergize with or counteract this modulation. Our findings indicate that increased membrane tension promotes the association of CCACs at integrin αVβ5 adhesion sites, leading to decreased cancer cell proliferation, spreading, and migration. Conversely, lower membrane tension promotes the formation of FAs, which correlates with the softer membranes observed in cancer cells, thus potentially facilitating cancer progression. Our research provides novel insights into the biomechanical regulation of CCACs and FAs, revealing their critical and contrasting roles in modulating cancer cell progression.
Collapse
Affiliation(s)
- Umida Djakbarova
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
| | - Yasaman Madraki
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
| | - Emily T. Chan
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Tianyao Wu
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | | | - A. Ata Akatay
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
| | - Comert Kural
- Department of Physics, The Ohio State University, Columbus, OH, 43210, USA
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
6
|
Gao X, Hou T, Wang L, Liu Y, Guo J, Zhang L, Yang T, Tang W, An M, Wen M. Aligned electrospun fibers of different diameters for improving cell migration capacity. Colloids Surf B Biointerfaces 2024; 234:113674. [PMID: 38039823 DOI: 10.1016/j.colsurfb.2023.113674] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/05/2023] [Revised: 10/27/2023] [Accepted: 11/24/2023] [Indexed: 12/03/2023]
Abstract
Electrospun fibers have gained significant attention as scaffolds in skin tissue engineering due to their biomimetic properties, which resemble the fibrous extracellular matrix. The morphological characteristics of electrospun fibers play a crucial role in determining cell behavior. However, the effects of electrospun fibers' arrangement and diameters on human skin fibroblasts (HSFs) remain elusive. Here, we revealed the impact of electrospun fiber diameters (700 nm, 2000 nm, and 3000 nm) on HSFs' proliferation, migration, and functional expression. The results demonstrated that all fibers exhibited good cytocompatibility. HSFs cultured on nanofibers (700 nm diameter) displayed a more dispersed and elongated morphology. Conversely, fibers with a diameter of 3000 nm exhibited a reduced specific surface area and lower adsorption of adhesion proteins, resulting in enhanced cell migration speed and effective migration rate. Meanwhile, the expression levels of migration-related genes and proteins were upregulated at 48 h for the 3000 nm fibers. This study demonstrated the unique role of fiber diameters in controlling the physiological functions of cells, especially decision-making and navigating migration in complex microenvironments of aligned electrospun fibers, and highlights the utility of these bioactive substitutes in skin tissue engineering applications.
Collapse
Affiliation(s)
- Xiang Gao
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China
| | - Tian Hou
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China
| | - Li Wang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China
| | - Yang Liu
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China
| | - Jiqiang Guo
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China
| | - Li Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Tiantian Yang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China
| | - Wenjie Tang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China
| | - Meiwen An
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China.
| | - Meiling Wen
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi Key Laboratory of Material Strength & Structural Impact, Taiyuan, Shanxi 030024, China.
| |
Collapse
|
7
|
Muhammed Y, Lazenby RA. Scanning ion conductance microscopy revealed cisplatin-induced morphological changes related to apoptosis in single adenocarcinoma cells. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:503-514. [PMID: 38167666 DOI: 10.1039/d3ay01827j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/05/2024]
Abstract
The studies of drug-induced apoptosis play a vital role in the identification of potential drugs that could treat diseases such as cancer. Alterations in the native morphology of cancer cells following treatment with anticancer drugs serve as one of the indicators that reveal drug efficacy. Various techniques such as optical microscopy, electron microscopy (EM), and atomic force microscopy (AFM) have been used to map the three dimensional (3D) morphological changes in cells induced with drugs. However, caution should be exercised when interpreting morphological data from techniques that might alter the native morphology of cells, caused by phototoxicity, electron beam invasiveness, intrusive sample preparation, and cell membrane deformation. Herein, we have used scanning ion conductance microscopy (SICM) to study the 3D morphology and roughness of A549 adenocarcinoma cells under physiological conditions before and after cisplatin induced apoptosis, where we observed an increase in height, overall shrinkage of the cells, and irregular features form on the cell membrane. Tracking the morphology of the same single A549 cells exposed to cisplatin unveiled heterogeneity in response to the drug, formation of membrane blebs, and an increase in membrane roughness. We have also demonstrated the use of SICM for studying the effect of cisplatin on the dynamic changes in the volume of A549 cells over days. SICM is demonstrated as a technique for studying the effect of drug induced apoptosis in the same cells over time, and for multiple different single cells.
Collapse
Affiliation(s)
- Yusuf Muhammed
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306-4390, USA.
| | - Robert A Lazenby
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, Florida 32306-4390, USA.
| |
Collapse
|
8
|
Cybulsky AV, Papillon J, Bryan C, Navarro‐Betancourt JR, Sabourin LA. Role of the Ste20-like kinase SLK in podocyte adhesion. Physiol Rep 2024; 12:e15897. [PMID: 38163671 PMCID: PMC10758337 DOI: 10.14814/phy2.15897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 01/03/2024] Open
Abstract
SLK controls the cytoskeleton, cell adhesion, and migration. Podocyte-specific deletion of SLK in mice leads to podocyte injury as mice age and exacerbates injury in experimental focal segment glomerulosclerosis (FSGS; adriamycin nephrosis). We hypothesized that adhesion proteins may be substrates of SLK. In adriamycin nephrosis, podocyte ultrastructural injury was exaggerated by SLK deletion. Analysis of a protein kinase phosphorylation site dataset showed that podocyte adhesion proteins-paxillin, vinculin, and talin-1 may be potential SLK substrates. In cultured podocytes, deletion of SLK increased adhesion to collagen. Analysis of paxillin, vinculin, and talin-1 showed that SLK deletion reduced focal adhesion complexes (FACs) containing these proteins mainly in adriamycin-induced injury; there was no change in FAC turnover (focal adhesion kinase Y397 phosphorylation). In podocytes, paxillin S250 showed basal phosphorylation that was slightly enhanced by SLK; however, SLK did not phosphorylate talin-1. In adriamycin nephrosis, SLK deletion did not alter glomerular expression/localization of talin-1 and vinculin, but increased focal adhesion kinase phosphorylation modestly. Therefore, SLK decreases podocyte adhesion, but FAC proteins in podocytes are not major substrates of SLK in health and disease.
Collapse
Affiliation(s)
- Andrey V. Cybulsky
- Department of MedicineMcGill University Health Centre Research Institute, McGill UniversityMontrealQuebecCanada
| | - Joan Papillon
- Department of MedicineMcGill University Health Centre Research Institute, McGill UniversityMontrealQuebecCanada
| | - Craig Bryan
- Department of MedicineMcGill University Health Centre Research Institute, McGill UniversityMontrealQuebecCanada
| | - José R. Navarro‐Betancourt
- Department of MedicineMcGill University Health Centre Research Institute, McGill UniversityMontrealQuebecCanada
| | - Luc A. Sabourin
- Ottawa Hospital Research Institute, Cancer TherapeuticsOttawaOntarioCanada
| |
Collapse
|
9
|
Biernacka Z, Gregorczyk-Zboroch K, Lasocka I, Ostrowska A, Struzik J, Gieryńska M, Toka FN, Szulc-Dąbrowska L. Ectromelia Virus Affects the Formation and Spatial Organization of Adhesive Structures in Murine Dendritic Cells In Vitro. Int J Mol Sci 2023; 25:558. [PMID: 38203729 PMCID: PMC10779027 DOI: 10.3390/ijms25010558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/29/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Ectromelia virus (ECTV) is a causative agent of mousepox. It provides a suitable model for studying the immunobiology of orthopoxviruses, including their interaction with the host cell cytoskeleton. As professional antigen-presenting cells, dendritic cells (DCs) control the pericellular environment, capture antigens, and present them to T lymphocytes after migration to secondary lymphoid organs. Migration of immature DCs is possible due to the presence of specialized adhesion structures, such as podosomes or focal adhesions (FAs). Since assembly and disassembly of adhesive structures are highly associated with DCs' immunoregulatory and migratory functions, we evaluated how ECTV infection targets podosomes and FAs' organization and formation in natural-host bone marrow-derived DCs (BMDC). We found that ECTV induces a rapid dissolution of podosomes at the early stages of infection, accompanied by the development of larger and wider FAs than in uninfected control cells. At later stages of infection, FAs were predominantly observed in long cellular extensions, formed extensively by infected cells. Dissolution of podosomes in ECTV-infected BMDCs was not associated with maturation and increased 2D cell migration in a wound healing assay; however, accelerated transwell migration of ECTV-infected cells towards supernatants derived from LPS-conditioned BMDCs was observed. We suggest that ECTV-induced changes in the spatial organization of adhesive structures in DCs may alter the adhesiveness/migration of DCs during some conditions, e.g., inflammation.
Collapse
Affiliation(s)
- Zuzanna Biernacka
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| | - Karolina Gregorczyk-Zboroch
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| | - Iwona Lasocka
- Department of Biology of Animal Environment, Institute of Animal Science, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Agnieszka Ostrowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland;
| | - Justyna Struzik
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| | - Małgorzata Gieryńska
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| | - Felix N. Toka
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre P.O. Box 334, Saint Kitts and Nevis
| | - Lidia Szulc-Dąbrowska
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences, 02-786 Warsaw, Poland; (Z.B.); (K.G.-Z.); (J.S.); (M.G.); (F.N.T.)
| |
Collapse
|
10
|
Nurmagambetova A, Mustyatsa V, Saidova A, Vorobjev I. Morphological and cytoskeleton changes in cells after EMT. Sci Rep 2023; 13:22164. [PMID: 38092761 PMCID: PMC10719275 DOI: 10.1038/s41598-023-48279-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/15/2023] [Accepted: 11/24/2023] [Indexed: 12/17/2023] Open
Abstract
Epithelial cells undergoing EMT experience significant alterations at transcriptional and morphological levels. However, changes in the cytoskeleton, especially cytoskeleton dynamics are poorly described. Addressing the question we induced EMT in three cell lines (MCF-7, HaCaT and A-549) and analyzed morphological and cytoskeletal changes there using immunostaining and life cell imaging of cells transfected with microtubule and focal adhesion markers. In all studied cell lines, cell area after EMT increased, MCF-7 and A-549 cells became elongated, while HaCaT cells kept the aspect ratio the same. We next analyzed three components of the cytoskeleton: microtubules, stress fibers and focal adhesions. The following changes were observed after EMT in cultured cells: (i) Organization of microtubules becomes more radial; and the growth rate of microtubule plus ends was accelerated; (ii) Actin stress fibers become co-aligned forming the longitudinal cell axis; and (iii) Focal adhesions had decreased area in all cancer cell lines studied and became more numerous in HaCaT cells. We conclude that among dynamic components of the cytoskeleton, the most significant changes during EMT happen in the regulation of microtubules.
Collapse
Affiliation(s)
- Assel Nurmagambetova
- School of Sciences and Humanities, Nazarbayev University, Kabanbay Batyr Avenue, 53, 010000, Astana, Kazakhstan.
- School of Engineering and Digital Sciences, Nazarbayev University, Kabanbay Batyr Avenue, 53, 010000, Astana, Kazakhstan.
| | - Vadim Mustyatsa
- National Laboratory Astana, Nazarbayev University, Kabanbay Batyr Avenue, 53, 010000, Astana, Kazakhstan
| | - Aleena Saidova
- National Laboratory Astana, Nazarbayev University, Kabanbay Batyr Avenue, 53, 010000, Astana, Kazakhstan
| | - Ivan Vorobjev
- School of Sciences and Humanities, Nazarbayev University, Kabanbay Batyr Avenue, 53, 010000, Astana, Kazakhstan.
- National Laboratory Astana, Nazarbayev University, Kabanbay Batyr Avenue, 53, 010000, Astana, Kazakhstan.
| |
Collapse
|
11
|
Ryan CN, Pugliese E, Shologu N, Gaspar D, Rooney P, Islam MN, O'Riordan A, Biggs MJ, Griffin MD, Zeugolis DI. Physicochemical cues are not potent regulators of human dermal fibroblast trans-differentiation. BIOMATERIALS AND BIOSYSTEMS 2023; 11:100079. [PMID: 37720487 PMCID: PMC10499661 DOI: 10.1016/j.bbiosy.2023.100079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/28/2022] [Revised: 04/25/2023] [Accepted: 05/29/2023] [Indexed: 09/19/2023] Open
Abstract
Due to their inherent plasticity, dermal fibroblasts hold great promise in regenerative medicine. Although biological signals have been well-established as potent regulators of dermal fibroblast function, it is still unclear whether physiochemical cues can induce dermal fibroblast trans-differentiation. Herein, we evaluated the combined effect of surface topography, substrate rigidity, collagen type I coating and macromolecular crowding in human dermal fibroblast cultures. Our data indicate that tissue culture plastic and collagen type I coating increased cell proliferation and metabolic activity. None of the assessed in vitro microenvironment modulators affected cell viability. Anisotropic surface topography induced bidirectional cell morphology, especially on more rigid (1,000 kPa and 130 kPa) substrates. Macromolecular crowding increased various collagen types, but not fibronectin, deposition. Macromolecular crowding induced globular extracellular matrix deposition, independently of the properties of the substrate. At day 14 (longest time point assessed), macromolecular crowding downregulated tenascin C (in 9 out of the 14 groups), aggrecan (in 13 out of the 14 groups), osteonectin (in 13 out of the 14 groups), and collagen type I (in all groups). Overall, our data suggest that physicochemical cues (such surface topography, substrate rigidity, collagen coating and macromolecular crowding) are not as potent as biological signals in inducing dermal fibroblast trans-differentiation.
Collapse
Affiliation(s)
- Christina N.M. Ryan
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Eugenia Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Naledi Shologu
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Peadar Rooney
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Md Nahidul Islam
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, University of Galway, Galway, Ireland
- Discipline of Biochemistry, School of Natural Sciences, University of Galway, Galway, Ireland
| | - Alan O'Riordan
- Tyndall National Institute, University College Cork (UCC), Cork, Ireland
| | - Manus J. Biggs
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Matthew D. Griffin
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
12
|
Hu P, Miller AE, Yeh CR, Bingham GC, Civelek M, Barker TH. SEMA7a primes integrin α5β1 engagement instructing fibroblast mechanotransduction, phenotype and transcriptional programming. Matrix Biol 2023; 121:179-193. [PMID: 37422024 DOI: 10.1016/j.matbio.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/18/2022] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023]
Abstract
Integrins are cellular receptors that bind the extracellular matrix (ECM) and facilitate the transduction of biochemical and biophysical microenvironment cues into cellular responses. Upon engaging the ECM, integrin heterodimers must rapidly strengthen their binding with the ECM, resulting in the assembly of force-resistant and force-sensitive integrin associated complexes (IACs). The IACs constitute an essential apparatus for downstream signaling and fibroblast phenotypes. During wound healing, integrin signaling is essential for fibroblast motility, proliferation, ECM reorganization and, ultimately, restoration of tissue homeostasis. Semaphorin 7A (SEMA7a) has been previously implicated in post-injury inflammation and tissue fibrosis, yet little is known about SEMA7a's role in directing stromal cell, particularly fibroblast, behaviors. We demonstrate that SEMA7a regulates integrin signaling through cis-coupling with active integrin α5β1 on the plasma membrane, enabling rapid integrin adhesion strengthening to fibronectin (Fn) and normal downstream mechanotransduction. This molecular function of SEMA7a potently regulates fibroblast adhesive, cytoskeletal, and migratory phenotype with strong evidence of downstream alterations in chromatin structure resulting in global transcriptomic reprogramming such that loss of SEMA7a expression is sufficient to impair the normal migratory and ECM assembly phenotype of fibroblasts resulting in significantly delayed tissue repair in vivo.
Collapse
Affiliation(s)
- Ping Hu
- Department of Biomedical Engineering, Schools of Engineering and Medicine, Charlottesville, VA 22908, USA
| | - Andrew E Miller
- Department of Biomedical Engineering, Schools of Engineering and Medicine, Charlottesville, VA 22908, USA
| | - Chiuan-Ren Yeh
- Department of Biomedical Engineering, Schools of Engineering and Medicine, Charlottesville, VA 22908, USA
| | - Grace C Bingham
- Department of Biomedical Engineering, Schools of Engineering and Medicine, Charlottesville, VA 22908, USA
| | - Mete Civelek
- Department of Biomedical Engineering, Schools of Engineering and Medicine, Charlottesville, VA 22908, USA; Center for Public Health Genomics, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Thomas H Barker
- Department of Biomedical Engineering, Schools of Engineering and Medicine, Charlottesville, VA 22908, USA.
| |
Collapse
|
13
|
Zbiral B, Weber A, Vivanco MDM, Toca-Herrera JL. Characterization of Breast Cancer Aggressiveness by Cell Mechanics. Int J Mol Sci 2023; 24:12208. [PMID: 37569585 PMCID: PMC10418463 DOI: 10.3390/ijms241512208] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/23/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
In healthy tissues, cells are in mechanical homeostasis. During cancer progression, this equilibrium is disrupted. Cancer cells alter their mechanical phenotype to a softer and more fluid-like one than that of healthy cells. This is connected to cytoskeletal remodeling, changed adhesion properties, faster cell proliferation and increased cell motility. In this work, we investigated the mechanical properties of breast cancer cells representative of different breast cancer subtypes, using MCF-7, tamoxifen-resistant MCF-7, MCF10A and MDA-MB-231 cells. We derived viscoelastic properties from atomic force microscopy force spectroscopy measurements and showed that the mechanical properties of the cells are associated with cancer cell malignancy. MCF10A are the stiffest and least fluid-like cells, while tamoxifen-resistant MCF-7 cells are the softest ones. MCF-7 and MDA-MB-231 show an intermediate mechanical phenotype. Confocal fluorescence microscopy on cytoskeletal elements shows differences in actin network organization, as well as changes in focal adhesion localization. These findings provide further evidence of distinct changes in the mechanical properties of cancer cells compared to healthy cells and add to the present understanding of the complex alterations involved in tumorigenesis.
Collapse
Affiliation(s)
- Barbara Zbiral
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (A.W.)
| | - Andreas Weber
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (A.W.)
| | - Maria dM. Vivanco
- Cancer Heterogeneity Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain;
| | - José L. Toca-Herrera
- Institute of Biophysics, Department of Bionanosciences, University of Natural Resources and Life Sciences, Muthgasse 11, 1190 Vienna, Austria; (B.Z.); (A.W.)
| |
Collapse
|
14
|
Chen HY, Wu YH, Wei CY, Liao ZY, Wu HT, Chen YC, Pang JHS. Incomplete Recovery from the Radiocontrast-Induced Dysregulated Cell Cycle, Adhesion, and Fibrogenesis in Renal Tubular Cells after Radiocontrast (Iohexol) Removal. Int J Mol Sci 2023; 24:10945. [PMID: 37446141 DOI: 10.3390/ijms241310945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/01/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Contrast-induced nephropathy (CIN) is one of the most common causes of acute kidney injury (AKI). However, management is still limited, and the cellular response to radiocontrast removal for CIN remains unclear. This study aimed to explore the latent effects of iohexol in cultured renal tubular cells with or without the removal of iohexol by medium replacement. HK2 renal tubular cells were subcultured 24 h before use in CIN experiments. Three treatment groups were established: the control, a radiocontrast (iohexol)-only group at 75 mg I/mL (I-75), and iohexol exposure for 24 h with culture medium replacement (I-75/M). Cell cycle arrest, fibrogenic mediator assays, cell viability, cell function, and cell-cycle-related protein expression were compared between groups. Iohexol induced numerous changes in HK2 renal tubular cells, such as enlarged cell shape, cell cycle arrest, increased apoptosis, and polyploidy. Iohexol inhibited the expression of cyclins, CDKs, ZO-1, and E-cadherin but conversely enhanced the expression of p21 and fibrosis-related genes, including TGF-β1, CTGF, collagen I, collagen III, and HIF-1α within 60 hr after the exposure. Except for the recovery from cell cycle arrest and cell cycle gene expression, notably, the removal of iohexol by medium replacement could not fully recover the renal tubular cells from the formation of polyploid cells, the adhesion or spreading, or the expression of fibrosis-related genes. The present study demonstrates, for the first time, that iohexol exerts latent cytotoxic effects on cultured renal tubular cells after its removal, suggesting that these irreversible cell changes may cause the insufficiency of radiocontrast reduction in CIN, which is worth investigating further.
Collapse
Affiliation(s)
- Hsing-Yu Chen
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Division of Chinese Internal Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33378, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yi-Hong Wu
- Division of Chinese Internal Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 33378, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Cheng-Yu Wei
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Zhi-Yao Liao
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Hsiao-Ting Wu
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yung-Chang Chen
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Division of Nephrology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan 33342, Taiwan
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, Taoyuan 33342, Taiwan
| |
Collapse
|
15
|
Chen J, Vishweshwaraiah YL, Mailman RB, Tabdanov ED, Dokholyan NV. A noncommutative combinatorial protein logic circuit controls cell orientation in nanoenvironments. SCIENCE ADVANCES 2023; 9:eadg1062. [PMID: 37235645 PMCID: PMC10219599 DOI: 10.1126/sciadv.adg1062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/02/2022] [Accepted: 04/20/2023] [Indexed: 05/28/2023]
Abstract
Single-protein-based devices that integrate signal sensing with logical operations to generate functional outputs offer exceptional promise for monitoring and modulating biological systems. Engineering such intelligent nanoscale computing agents is challenging, as it requires the integration of sensor domains into a functional protein via intricate allosteric networks. We incorporate a rapamycin-sensitive sensor (uniRapR) and a blue light-responsive LOV2 domain into human Src kinase, creating a protein device that functions as a noncommutative combinatorial logic circuit. In our design, rapamycin activates Src kinase, causing protein localization to focal adhesions, whereas blue light exerts the reverse effect that inactivates Src translocation. Focal adhesion maturation induced by Src activation reduces cell migration dynamics and shifts cell orientation to align along collagen nanolane fibers. Using this protein device, we reversibly control cell orientation by applying the appropriate input signals, a framework that may be useful in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Jiaxing Chen
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033-0850, USA
| | | | - Richard B. Mailman
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033-0850, USA
| | - Erdem D. Tabdanov
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033-0850, USA
| | - Nikolay V. Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033-0850, USA
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA 17033-0850, USA
- Department of Chemistry, Pennsylvania State University, University Park, PA 16802, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
16
|
Riivari S, Närvä E, Kangasniemi I, Willberg J, Närhi T. Focal adhesion formation of primary human gingival fibroblast on hydrothermally and in-sol-made TiO 2 -coated titanium. Clin Implant Dent Relat Res 2023. [PMID: 36815407 DOI: 10.1111/cid.13195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/21/2022] [Revised: 01/31/2023] [Accepted: 02/11/2023] [Indexed: 02/24/2023]
Abstract
Optimal cell adhesion of the gingival fibroblasts to dental implants is important for maintaining good implant integration. The aim of this study was to discover, if the nanoporous TiO2 -coating on titanium alloy substrates is able to increase the cell adhesion of the human gingival fibroblasts (HGF). The study consisted of three differently produced titanium groups: hydrothermally produced TiO2 -coating (HT), novel TiO2 -coating made in sol (SOL), and noncoated control group. Primary HGF cells were initiated from gingival biopsies from patients having a third molar extraction. HGF were cultivated on titanium discs for 2 and 24 h to determine the initial attachment with confocal microscope. The cell spreading and adhesion protein signals were measured. In addition, expression of adhesion proteins vinculin, paxillin, and focal adhesion kinase (FAK) were measured after 3 days of cultivation by using Western Blotting. Higher protein levels of paxillin, vinculin, and FAK were induced on both coated discs compared to noncoated discs. The difference was statistically significant (p < 0.05) concerning expression of paxillin. The cell spreading was significantly larger on SOL discs after 2 and 24 h when comparing to noncoated controls. The confocal microscope analyses revealed significantly higher adhesion protein signals on both HT- and SOL-coated titanium compared to control group. This study showed, that both methods to produce TiO2 -coatings are able to increase HGF adhesion protein expression and cell spreading on titanium surface. Accordingly, the coatings can potentially improve the gingival attachment to titanium implant surfaces.
Collapse
Affiliation(s)
- Sini Riivari
- Department of Prosthetic Dentistry and Stomatognathic Physiology, University of Turku, Turku, Finland
| | - Elisa Närvä
- Institute of Biomedicine and Cancer Research Laboratory FICAN West, University of Turku, Turku, Finland
| | | | - Jaana Willberg
- Department of Oral Pathology and Oral Radiology, University of Turku, Turku, Finland.,Department of Pathology, Turku University Central Hospital, Turku, Finland
| | - Timo Närhi
- Department of Prosthetic Dentistry and Stomatognathic Physiology, University of Turku, Turku, Finland
| |
Collapse
|
17
|
Ryan CNM, Pugliese E, Shologu N, Gaspar D, Rooney P, Islam MN, O'Riordan A, Biggs MJ, Griffin MD, Zeugolis DI. The synergistic effect of physicochemical in vitro microenvironment modulators in human bone marrow stem cell cultures. BIOMATERIALS ADVANCES 2022; 144:213196. [PMID: 36455498 DOI: 10.1016/j.bioadv.2022.213196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 07/11/2022] [Revised: 10/29/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Modern bioengineering utilises biomimetic cell culture approaches to control cell fate during in vitro expansion. In this spirit, herein we assessed the influence of bidirectional surface topography, substrate rigidity, collagen type I coating and macromolecular crowding (MMC) in human bone marrow stem cell cultures. In the absence of MMC, surface topography was a strong modulator of cell morphology. MMC significantly increased extracellular matrix deposition, albeit in a globular manner, independently of the surface topography, substrate rigidity and collagen type I coating. Collagen type I coating significantly increased cell metabolic activity and none of the assessed parameters affected cell viability. At day 14, in the absence of MMC, none of the assessed genes was affected by surface topography, substrate rigidity and collagen type I coating, whilst in the presence of MMC, in general, collagen type I α1 chain, tenascin C, osteonectin, bone sialoprotein, aggrecan, cartilage oligomeric protein and runt-related transcription factor were downregulated. Interestingly, in the presence of the MMC, the 1000 kPa grooved substrate without collagen type I coating upregulated aggrecan, cartilage oligomeric protein, scleraxis homolog A, tenomodulin and thrombospondin 4, indicative of tenogenic differentiation. This study further supports the notion for multifactorial bioengineering to control cell fate in culture.
Collapse
Affiliation(s)
- Christina N M Ryan
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Eugenia Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Naledi Shologu
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Peadar Rooney
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Md Nahidul Islam
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Discipline of Biochemistry, School of Natural Sciences, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Alan O'Riordan
- Tyndall National Institute, University College Cork (UCC), Cork, Ireland
| | - Manus J Biggs
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Matthew D Griffin
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
18
|
Harati J, Tao X, Shahsavarani H, Du P, Galluzzi M, Liu K, Zhang Z, Shaw P, Shokrgozar MA, Pan H, Wang PY. Polydopamine-Mediated Protein Adsorption Alters the Epigenetic Status and Differentiation of Primary Human Adipose-Derived Stem Cells (hASCs). Front Bioeng Biotechnol 2022; 10:934179. [PMID: 36032703 PMCID: PMC9399727 DOI: 10.3389/fbioe.2022.934179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/02/2022] [Accepted: 06/08/2022] [Indexed: 11/20/2022] Open
Abstract
Polydopamine (PDA) is a biocompatible cell-adhesive polymer with versatile applications in biomedical devices. Previous studies have shown that PDA coating could improve cell adhesion and differentiation of human mesenchymal stem cells (hMSCs). However, there is still a knowledge gap in the effect of PDA-mediated protein adsorption on the epigenetic status of MSCs. This work used gelatin-coated cell culture surfaces with and without PDA underlayer (Gel and PDA-Gel) to culture and differentiate primary human adipose-derived stem cells (hASCs). The properties of these two substrates were significantly different, which, in combination with a variation in extracellular matrix (ECM) protein bioactivity, regulated cell adhesion and migration. hASCs reduced focal adhesions by downregulating the expression of integrins such as αV, α1, α2, and β1 on the PDA-Gel compared to the Gel substrate. Interestingly, the ratio of H3K27me3 to H3K27me3+H3K4me3 was decreased, but this only occurred for upregulation of AGG and BMP4 genes during chondrogenic differentiation. This result implies that the PDA-Gel surface positively affects the chondrogenic, but not adipogenic and osteogenic, differentiation. In conclusion, for the first time, this study demonstrates the sequential effects of PDA coating on the biophysical property of adsorbed protein and then focal adhesions and differentiation of hMSCs through epigenetic regulation. This study sheds light on PDA-mediated mechanotransduction.
Collapse
Affiliation(s)
- Javad Harati
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
- Lab Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Xuelian Tao
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Ping Du
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Massimiliano Galluzzi
- Materials Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Kun Liu
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Zhang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Peter Shaw
- Oujiang Laboratory, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Mohammad Ali Shokrgozar
- Lab Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | - Haobo Pan
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- *Correspondence: Peng-Yuan Wang, ; Haobo Pan,
| | - Peng-Yuan Wang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Oujiang Laboratory, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
- *Correspondence: Peng-Yuan Wang, ; Haobo Pan,
| |
Collapse
|
19
|
Allahyari Z, Gaborski TR. Engineering cell-substrate interactions on porous membranes for microphysiological systems. LAB ON A CHIP 2022; 22:2080-2089. [PMID: 35593461 DOI: 10.1039/d2lc00114d] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/15/2023]
Abstract
Microphysiological systems are now widely used to recapitulate physiological and pathological microenvironments in order to study and understand a variety of cellular processes as well as drug delivery and stem cell differentiation. Central to many of these systems are porous membranes that enable tissue barrier formation as well as compartmentalization while still facilitating small molecule diffusion, cellular transmigration and cell-cell communication. The role or impact of porous membranes on the cells cultured upon them has not been widely studied or reviewed. Although many chemical and physical substrate characteristics have been shown to be effective in controlling and directing cellular behavior, the influence of pore characteristics and the ability to engineer porous membranes to influence these responses is not fully understood. In this mini-review, we show that many studies point to a multiphasic cell-substrate response, where increasing pore sizes and pore-pore spacing generally leads to improved cell-substrate interactions. However, the smallest pores in the nano-scale sometimes promote the strongest cell-substrate interactions, while the very largest micron-scale pores hinder cell-substrate interactions. This synopsis provides an insight into the importance of membrane pores in controlling cellular responses, and may help with the design and utilization of porous membranes for induction of desired cell processes in the development of biomimetic platforms.
Collapse
Affiliation(s)
- Zahra Allahyari
- Department of Microsystems Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, NY 14623, USA.
- Department of Biomedical Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, NY 14623, USA
| | - Thomas R Gaborski
- Department of Microsystems Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, NY 14623, USA.
- Department of Biomedical Engineering, Rochester Institute of Technology, 160 Lomb Memorial Drive, Rochester, NY 14623, USA
| |
Collapse
|
20
|
Lehne F, Pokrant T, Parbin S, Salinas G, Großhans J, Rust K, Faix J, Bogdan S. Calcium bursts allow rapid reorganization of EFhD2/Swip-1 cross-linked actin networks in epithelial wound closure. Nat Commun 2022; 13:2492. [PMID: 35524157 PMCID: PMC9076686 DOI: 10.1038/s41467-022-30167-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/26/2021] [Accepted: 04/19/2022] [Indexed: 02/01/2023] Open
Abstract
Changes in cell morphology require the dynamic remodeling of the actin cytoskeleton. Calcium fluxes have been suggested as an important signal to rapidly relay information to the actin cytoskeleton, but the underlying mechanisms remain poorly understood. Here, we identify the EF-hand domain containing protein EFhD2/Swip-1 as a conserved lamellipodial protein strongly upregulated in Drosophila macrophages at the onset of metamorphosis when macrophage behavior shifts from quiescent to migratory state. Loss- and gain-of-function analysis confirm a critical function of EFhD2/Swip-1 in lamellipodial cell migration in fly and mouse melanoma cells. Contrary to previous assumptions, TIRF-analyses unambiguously demonstrate that EFhD2/Swip-1 proteins efficiently cross-link actin filaments in a calcium-dependent manner. Using a single-cell wounding model, we show that EFhD2/Swip-1 promotes wound closure in a calcium-dependent manner. Mechanistically, our data suggest that transient calcium bursts reduce EFhD2/Swip-1 cross-linking activity and thereby promote rapid reorganization of existing actin networks to drive epithelial wound closure.
Collapse
Affiliation(s)
- Franziska Lehne
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University Marburg, Marburg, Germany
| | - Thomas Pokrant
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Sabnam Parbin
- NGS-Integrative Genomics Core Unit, Department of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit, Department of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Jörg Großhans
- Department of Biology, Philipps-University Marburg, Marburg, Germany
| | - Katja Rust
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University Marburg, Marburg, Germany
| | - Jan Faix
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Sven Bogdan
- Institute of Physiology and Pathophysiology, Department of Molecular Cell Physiology, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
21
|
Xia J, Liu ZY, Han ZY, Yuan Y, Shao Y, Feng XQ, Weitz DA. Regulation of cell attachment, spreading, and migration by hydrogel substrates with independently tunable mesh size. Acta Biomater 2022; 141:178-189. [PMID: 35041902 PMCID: PMC8898306 DOI: 10.1016/j.actbio.2022.01.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/09/2021] [Revised: 12/25/2021] [Accepted: 01/13/2022] [Indexed: 12/12/2022]
Abstract
Hydrogels are widely used as substrates to investigate interactions between cells and their microenvironment as they mimic many attributes of the extracellular matrix. The stiffness of hydrogels is an important property that is known to regulate cell behavior. Beside stiffness, cells also respond to structural cues such as mesh size. However, since the mesh size of hydrogel is intrinsically coupled to its stiffness, its role in regulating cell behavior has never been independently investigated. Here, we report a hydrogel system whose mesh size and stiffness can be independently controlled. Cell behavior, including spreading, migration, and formation of focal adhesions is significantly altered on hydrogels with different mesh sizes but with the same stiffness. At the transcriptional level, hydrogel mesh size affects cellular mechanotransduction by regulating nuclear translocation of yes-associated protein. These findings demonstrate that the mesh size of a hydrogel plays an important role in cell-substrate interactions. STATEMENT OF SIGNIFICANCE: Hydrogels are ideal platforms with which to investigate interactions between cells and their microenvironment as they mimic many physical properties of the extracellular matrix. However, the mesh size of hydrogels is intrinsically coupled to their stiffness, making it challenging to investigate the contribution of mesh size to cell behavior. In this work, we use hydrogel-on-glass substrates with defined thicknesses whose stiffness and mesh size can be independently tuned. We use these substrates to isolate the effects of mesh size on cell behavior, including attachment, spreading, migration, focal adhesion formation and YAP localization in the nucleus. Our results show that mesh size has significant, yet often overlooked, effects, on cell behavior, and contribute to a further understanding of cell-substrate interactions.
Collapse
Affiliation(s)
- Jing Xia
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Zong-Yuan Liu
- Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing 100084, China
| | - Zheng-Yuan Han
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Yuan Yuan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
| | - Yue Shao
- Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing 100084, China
| | - Xi-Qiao Feng
- Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing 100084, China.
| | - David A Weitz
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Department of Physics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
22
|
Roblek M, Bicher J, van Gogh M, György A, Seeböck R, Szulc B, Damme M, Olczak M, Borsig L, Siekhaus DE. The Solute Carrier MFSD1 Decreases the Activation Status of β1 Integrin and Thus Tumor Metastasis. Front Oncol 2022; 12:777634. [PMID: 35211397 PMCID: PMC8861502 DOI: 10.3389/fonc.2022.777634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/15/2021] [Accepted: 01/13/2022] [Indexed: 11/17/2022] Open
Abstract
Solute carriers are increasingly recognized as participating in a plethora of pathologies, including cancer. We describe here the involvement of the orphan solute carrier Major Facilitator Superfamily Domain-containing protein 1 (MFSD1) in the regulation of tumor cell migration. Loss of MFSD1 enabled higher levels of metastasis in experimental and spontaneous metastasis mouse models. We identified an increased migratory potential in MFSD1−/− tumor cells which was mediated by increased focal adhesion turnover, reduced stability of mature inactive β1 integrin, and the resulting increased integrin activation index. We show that MFSD1 promoted recycling to the cell surface of endocytosed inactive β1 integrin and thereby protected β1 integrin from proteolytic degradation; this led to dampening of the integrin activation index. Furthermore, downregulation of MFSD1 expression was observed during the early steps of tumorigenesis, and higher MFSD1 expression levels correlate with a better cancer patient prognosis. In sum, we describe a requirement for endolysosomal MFSD1 in efficient β1 integrin recycling to suppress tumor cell dissemination.
Collapse
Affiliation(s)
- Marko Roblek
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Julia Bicher
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Merel van Gogh
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Attila György
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Rita Seeböck
- Institute of Clinical Pathology, University Hospital St. Polten, St. Polten, Austria
| | - Bozena Szulc
- Laboratory of Biochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Markus Damme
- Institute of Biochemistry, University of Kiel, Kiel, Germany
| | - Mariusz Olczak
- Laboratory of Biochemistry, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Lubor Borsig
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Daria E Siekhaus
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| |
Collapse
|
23
|
Modulation of stem cell response using biodegradable polyester films with different stiffness. BIOMEDICAL ENGINEERING ADVANCES 2021. [DOI: 10.1016/j.bea.2021.100007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022] Open
|
24
|
Cacopardo L, Guazzelli N, Ahluwalia A. Characterising and engineering biomimetic materials for viscoelastic mechanotransduction studies. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:912-925. [PMID: 34555953 PMCID: PMC9419958 DOI: 10.1089/ten.teb.2021.0151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Indexed: 11/24/2022]
Abstract
The mechanical behavior of soft tissue extracellular matrix is time dependent. Moreover, it evolves over time due to physiological processes as well as aging and disease. Measuring and quantifying the time-dependent mechanical behavior of soft tissues and materials pose a challenge, not only because of their labile and hydrated nature but also because of the lack of a common definition of terms and understanding of models for characterizing viscoelasticity. Here, we review the most important measurement techniques and models used to determine the viscoelastic properties of soft hydrated materials—or hydrogels—underlining the difference between viscoelastic behavior and the properties and descriptors used to quantify viscoelasticity. We then discuss the principal factors, which determine tissue viscoelasticity in vivo and summarize what we currently know about cell response to time-dependent materials, outlining fundamental factors that have to be considered when interpreting results. Particular attention is given to the relationship between the different time scales involved (mechanical, cellular and observation time scales), as well as scaling principles, all of which must be considered when designing viscoelastic materials and performing experiments for biomechanics or mechanobiology applications. From this overview, key considerations and directions for furthering insights and applications in the emergent field of cell viscoelastic mechanotransduction are provided.
Collapse
Affiliation(s)
| | - Nicole Guazzelli
- University of Pisa, 9310, Research Center 'E.Piaggio', Pisa, Italy.,University of Pisa, 9310, Information Engineering Department, Pisa, Italy;
| | - Arti Ahluwalia
- University of Pisa, 9310, Pisa, Italy.,University of Pisa, 9310, Information Engineering Department, Pisa, Toscana, Italy.,Centro 3R (Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research), Pisa, Italy;
| |
Collapse
|
25
|
Ryan C, Pugliese E, Shologu N, Gaspar D, Rooney P, Islam MN, O'Riordan A, Biggs M, Griffin M, Zeugolis D. A combined physicochemical approach towards human tenocyte phenotype maintenance. Mater Today Bio 2021; 12:100130. [PMID: 34632361 PMCID: PMC8488312 DOI: 10.1016/j.mtbio.2021.100130] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 02/08/2023] Open
Abstract
During in vitro culture, bereft of their optimal tissue context, tenocytes lose their phenotype and function. Considering that tenocytes in their native tissue milieu are exposed simultaneously to manifold signals, combination approaches (e.g. growth factor supplementation and mechanical stimulation) are continuously gaining pace to control cell fate during in vitro expansion, albeit with limited success due to the literally infinite number of possible permutations. In this work, we assessed the potential of scalable and potent physicochemical approaches that control cell fate (substrate stiffness, anisotropic surface topography, collagen type I coating) and enhance extracellular matrix deposition (macromolecular crowding) in maintaining human tenocyte phenotype in culture. Cell morphology was primarily responsive to surface topography. The tissue culture plastic induced the largest nuclei area, the lowest aspect ratio, and the highest focal adhesion kinase. Collagen type I coating increased cell number and metabolic activity. Cell viability was not affected by any of the variables assessed. Macromolecular crowding intensely enhanced and accelerated native extracellular matrix deposition, albeit not in an aligned fashion, even on the grooved substrates. Gene analysis at day 14 revealed that the 130 kPa grooved substrate without collagen type I coating and under macromolecular crowding conditions positively regulated human tenocyte phenotype. Collectively, this work illustrates the beneficial effects of combined physicochemical approaches in controlling cell fate during in vitro expansion.
Collapse
Affiliation(s)
- C.N.M. Ryan
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - E. Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - N. Shologu
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - D. Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - P. Rooney
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Md N. Islam
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Discipline of Biochemistry, School of Natural Sciences, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - A. O'Riordan
- Tyndall National Institute, University College Cork (UCC), Cork, Ireland
| | - M.J. Biggs
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - M.D. Griffin
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Regenerative Medicine Institute (REMEDI), School of Medicine, Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - D.I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
26
|
Adebowale K, Gong Z, Hou JC, Wisdom KM, Garbett D, Lee HP, Nam S, Meyer T, Odde DJ, Shenoy VB, Chaudhuri O. Enhanced substrate stress relaxation promotes filopodia-mediated cell migration. NATURE MATERIALS 2021; 20:1290-1299. [PMID: 33875851 PMCID: PMC8390443 DOI: 10.1038/s41563-021-00981-w] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/20/2019] [Accepted: 03/09/2021] [Indexed: 05/11/2023]
Abstract
Cell migration on two-dimensional substrates is typically characterized by lamellipodia at the leading edge, mature focal adhesions and spread morphologies. These observations result from adherent cell migration studies on stiff, elastic substrates, because most cells do not migrate on soft, elastic substrates. However, many biological tissues are soft and viscoelastic, exhibiting stress relaxation over time in response to a deformation. Here, we have systematically investigated the impact of substrate stress relaxation on cell migration on soft substrates. We observed that cells migrate minimally on substrates with an elastic modulus of 2 kPa that are elastic or exhibit slow stress relaxation, but migrate robustly on 2-kPa substrates that exhibit fast stress relaxation. Strikingly, migrating cells were not spread out and did not extend lamellipodial protrusions, but were instead rounded, with filopodia protrusions extending at the leading edge, and exhibited small nascent adhesions. Computational models of cell migration based on a motor-clutch framework predict the observed impact of substrate stress relaxation on cell migration and filopodia dynamics. Our findings establish substrate stress relaxation as a key requirement for robust cell migration on soft substrates and uncover a mode of two-dimensional cell migration marked by round morphologies, filopodia protrusions and weak adhesions.
Collapse
Affiliation(s)
- Kolade Adebowale
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Ze Gong
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jay C Hou
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Katrina M Wisdom
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Damien Garbett
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hong-Pyo Lee
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Sungmin Nam
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | - Tobias Meyer
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Vivek B Shenoy
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Ovijit Chaudhuri
- Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
27
|
Osuchowska PN, Wachulak P, Kasprzycka W, Nowak-Stępniowska A, Wakuła M, Bartnik A, Fiedorowicz H, Trafny EA. Adhesion of Triple-Negative Breast Cancer Cells under Fluorescent and Soft X-ray Contact Microscopy. Int J Mol Sci 2021; 22:ijms22147279. [PMID: 34298899 PMCID: PMC8306697 DOI: 10.3390/ijms22147279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/31/2021] [Revised: 06/24/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022] Open
Abstract
Understanding cancer cell adhesion could help to diminish tumor progression and metastasis. Adhesion mechanisms are currently the main therapeutic target of TNBC-resistant cells. This work shows the distribution and size of adhesive complexes determined with a common fluorescence microscopy technique and soft X-ray contact microscopy (SXCM). The results presented here demonstrate the potential of applying SXCM for imaging cell protrusions with high resolution when the cells are still alive in a physiological buffer. The possibility to observe the internal components of cells at a pristine and hydrated state with nanometer resolution distinguishes SXCM from the other more commonly used techniques for cell imaging. Thus, SXCM can be a promising technique for investigating the adhesion and organization of the actin cytoskeleton in cancer cells.
Collapse
Affiliation(s)
- Paulina Natalia Osuchowska
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Kaliskiego 2, 00-908 Warsaw, Poland; (P.N.O.); (W.K.); (A.N.-S.)
| | - Przemysław Wachulak
- Laser Technology Division, Institute of Optoelectronics, Military University of Technology, Kaliskiego 2, 00-908 Warsaw, Poland; (P.W.); (A.B.)
| | - Wiktoria Kasprzycka
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Kaliskiego 2, 00-908 Warsaw, Poland; (P.N.O.); (W.K.); (A.N.-S.)
| | - Agata Nowak-Stępniowska
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Kaliskiego 2, 00-908 Warsaw, Poland; (P.N.O.); (W.K.); (A.N.-S.)
| | - Maciej Wakuła
- Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.W.); (H.F.)
| | - Andrzej Bartnik
- Laser Technology Division, Institute of Optoelectronics, Military University of Technology, Kaliskiego 2, 00-908 Warsaw, Poland; (P.W.); (A.B.)
| | - Henryk Fiedorowicz
- Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland; (M.W.); (H.F.)
| | - Elżbieta Anna Trafny
- Biomedical Engineering Centre, Institute of Optoelectronics, Military University of Technology, Kaliskiego 2, 00-908 Warsaw, Poland; (P.N.O.); (W.K.); (A.N.-S.)
- Correspondence: ; Tel.: +48-261-839-544
| |
Collapse
|
28
|
Rose M, Noetzel E, Kistermann J, Eschenbruch J, Rushrush S, Gan L, Knüchel R, Gaisa NT, Dahl E. The ECM Modulator ITIH5 Affects Cell Adhesion, Motility and Chemotherapeutic Response of Basal/Squamous-Like (BASQ) Bladder Cancer Cells. Cells 2021; 10:cells10051038. [PMID: 33924987 PMCID: PMC8146567 DOI: 10.3390/cells10051038] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/17/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 12/16/2022] Open
Abstract
This study aims at characterizing the role of the putative tumor suppressor ITIH5 in basal-type bladder cancers (BLCA). By sub-classifying TCGA BLCA data, we revealed predominant loss of ITIH5 expression in the basal/squamous-like (BASQ) subtype. ITIH5 expression inversely correlated with basal-type makers such as KRT6A and CD44. Interestingly, Kaplan–Meier analyses showed longer recurrence-free survival in combination with strong CD44 expression, which is thought to mediate ITIH-hyaluronan (HA) binding functions. In vitro, stable ITIH5 overexpression in two basal-type BLCA cell lines showing differential CD44 expression levels, i.e., with (SCaBER) and without squamous features (HT1376), demonstrated clear inhibition of cell and colony growth of BASQ-type SCaBER cells. ITIH5 further enhanced HA-associated cell-matrix attachment, indicated by altered size and number of focal adhesion sites resulting in reduced cell migration capacities. Transcriptomic analyses revealed enrichment of pathways and processes involved in ECM organization, differentiation and cell signaling. Finally, we provide evidence that ITIH5 increase sensitivity of SCaBER cells to chemotherapeutical agents (cisplatin and gemcitabine), whereas responsiveness of HT1376 cells was not affected by ITIH5 expression. Thus, we gain further insights into the putative role of ITIH5 as tumor suppressor highlighting an impact on drug response potentially via the HA-CD44 axis in BASQ-type BLCA.
Collapse
MESH Headings
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Adhesion
- Cell Proliferation
- Cisplatin/administration & dosage
- DNA Methylation
- Deoxycytidine/administration & dosage
- Deoxycytidine/analogs & derivatives
- Gene Expression Regulation, Neoplastic
- Humans
- Neoplasms, Basal Cell/drug therapy
- Neoplasms, Basal Cell/genetics
- Neoplasms, Basal Cell/metabolism
- Neoplasms, Basal Cell/pathology
- Prognosis
- Promoter Regions, Genetic
- Proteinase Inhibitory Proteins, Secretory/genetics
- Proteinase Inhibitory Proteins, Secretory/metabolism
- Retrospective Studies
- Survival Rate
- Tumor Cells, Cultured
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/pathology
- Gemcitabine
Collapse
Affiliation(s)
- Michael Rose
- Institute of Pathology, University Hospital RWTH Aachen University, 52074 Aachen, Germany; (J.K.); (S.R.); (R.K.); (N.T.G.)
- Correspondence: (M.R.); (E.D.); Tel.: +49-241-80-89715 (M.R.); +49-241-80-88431 (E.D.); Fax: +49-241-8082439 (M.R. & E.D.)
| | - Erik Noetzel
- Institute of Biological Information Processing 2 (IBI-2), Mechanobiology, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany; (E.N.); (J.E.)
| | - Jennifer Kistermann
- Institute of Pathology, University Hospital RWTH Aachen University, 52074 Aachen, Germany; (J.K.); (S.R.); (R.K.); (N.T.G.)
| | - Julian Eschenbruch
- Institute of Biological Information Processing 2 (IBI-2), Mechanobiology, Forschungszentrum Jülich GmbH, 52425 Jülich, Germany; (E.N.); (J.E.)
| | - Sandra Rushrush
- Institute of Pathology, University Hospital RWTH Aachen University, 52074 Aachen, Germany; (J.K.); (S.R.); (R.K.); (N.T.G.)
| | - Lin Gan
- IZKF Aachen, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany;
| | - Ruth Knüchel
- Institute of Pathology, University Hospital RWTH Aachen University, 52074 Aachen, Germany; (J.K.); (S.R.); (R.K.); (N.T.G.)
| | - Nadine T. Gaisa
- Institute of Pathology, University Hospital RWTH Aachen University, 52074 Aachen, Germany; (J.K.); (S.R.); (R.K.); (N.T.G.)
| | - Edgar Dahl
- Institute of Pathology, University Hospital RWTH Aachen University, 52074 Aachen, Germany; (J.K.); (S.R.); (R.K.); (N.T.G.)
- Correspondence: (M.R.); (E.D.); Tel.: +49-241-80-89715 (M.R.); +49-241-80-88431 (E.D.); Fax: +49-241-8082439 (M.R. & E.D.)
| |
Collapse
|
29
|
Ciriza J, Rodríguez-Romano A, Nogueroles I, Gallego-Ferrer G, Cabezuelo RM, Pedraz JL, Rico P. Borax-loaded injectable alginate hydrogels promote muscle regeneration in vivo after an injury. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 123:112003. [PMID: 33812623 PMCID: PMC8085734 DOI: 10.1016/j.msec.2021.112003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 10/30/2020] [Revised: 02/05/2021] [Accepted: 02/20/2021] [Indexed: 11/25/2022]
Abstract
Muscle tissue possess an innate regenerative potential that involves an extremely complicated and synchronized process on which resident muscle stem cells play a major role: activate after an injury, differentiate and fuse originating new myofibers for muscle repair. Considerable efforts have been made to design new approaches based on material systems to potentiate muscle repair by engineering muscle extracellular matrix and/or including soluble factors/cells in the media, trying to recapitulate the key biophysical and biochemical cues present in the muscle niche. This work proposes a different and simple approach to potentiate muscle regeneration exploiting the interplay between specific cell membrane receptors. The simultaneous stimulation of borate transporter, NaBC1 (encoded by SLC4A11gene), and fibronectin-binding integrins induced higher number and size of focal adhesions, major cell spreading and actin stress fibers, strengthening myoblast attachment and providing an enhanced response in terms of myotube fusion and maturation. The stimulated NaBC1 generated an adhesion-driven state through a mechanism that involves simultaneous NaBC1/α5β1/αvβ3 co-localization. We engineered and characterized borax-loaded alginate hydrogels for an effective activation of NaBC1 in vivo. After inducing an acute injury with cardiotoxin in mice, active-NaBC1 accelerated the muscle regeneration process. Our results put forward a new biomaterial approach for muscle repair.
Collapse
Affiliation(s)
- Jesús Ciriza
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain; NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, C/ Miguel de Unamuno, 3, 01006 Vitoria Gasteiz, Spain.
| | - Ana Rodríguez-Romano
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain; Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Ignacio Nogueroles
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain
| | - Gloria Gallego-Ferrer
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain; Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| | - Rubén Martín Cabezuelo
- Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| | - José Luis Pedraz
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain; NanoBioCel Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country UPV/EHU, C/ Miguel de Unamuno, 3, 01006 Vitoria Gasteiz, Spain.
| | - Patricia Rico
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain; Center for Biomaterials and Tissue Engineering (CBIT), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| |
Collapse
|
30
|
Campos FSO, Piña-Rodrigues FM, Reis A, Atella GC, Mermelstein CS, Allodi S, Cavalcante LA. Lipid Rafts from Olfactory Ensheathing Cells: Molecular Composition and Possible Roles. Cell Mol Neurobiol 2021; 41:525-536. [PMID: 32415577 DOI: 10.1007/s10571-020-00869-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/13/2020] [Accepted: 05/05/2020] [Indexed: 01/16/2023]
Abstract
Olfactory ensheathing cells (OECs) are specialized glial cells of the olfactory system, believed to play a role in the continuous production of olfactory neurons and ensheathment of their axons. Although OECs are used in therapeutic applications, little is known about the cellular mechanisms underlying their migratory behavior. Recently, we showed that OEC migration is sensitive to ganglioside blockage through A2B5 and Jones antibody in OEC culture. Gangliosides are common components of lipid rafts, where they participate in several cellular mechanisms, including cell migration. Here, we characterized OEC lipid rafts, analyzing the presence of specific proteins and gangliosides that are commonly expressed in motile neural cells, such as young neurons, oligodendrocyte progenitors, and glioma cells. Our results showed that lipid rafts isolated from OECs were enriched in cholesterol, sphingolipids, phosphatidylcholine, caveolin-1, flotillin-1, gangliosides GM1 and 9-O-acetyl GD3, A2B5-recognized gangliosides, CNPase, α-actinin, and β1-integrin. Analysis of the actin cytoskeleton of OECs revealed stress fibers, membrane spikes, ruffled membranes and lamellipodia during cell migration, as well as the distribution of α-actinin in membrane projections. This is the first description of α-actinin and flotillin-1 in lipid rafts isolated from OECs and suggests that, together with β1-integrin and gangliosides, membrane lipid rafts play a role during OEC migration. This study provides new information on the molecular composition of OEC membrane microdomains that can impact on our understanding of the role of OEC lipid rafts under physiological and pathological conditions of the nervous system, including inflammation, hypoxia, aging, neurodegenerative diseases, head trauma, brain tumor, and infection.
Collapse
Affiliation(s)
- Fernanda S O Campos
- Laboratório de Neurobiologia do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, CCS. Bloco G, Rio de Janeiro, 21941-902, Brazil
- Laboratório de Bioquímica de Lipídeos e Lipoproteínas, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Felipe M Piña-Rodrigues
- Laboratório de Neurobiologia do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, CCS. Bloco G, Rio de Janeiro, 21941-902, Brazil
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Alice Reis
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Georgia C Atella
- Laboratório de Bioquímica de Lipídeos e Lipoproteínas, Instituto de Bioquímica Médica, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Claudia S Mermelstein
- Laboratório de Diferenciação Muscular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Silvana Allodi
- Laboratório de Neurobiologia do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, CCS. Bloco G, Rio de Janeiro, 21941-902, Brazil.
| | - Leny A Cavalcante
- Laboratório de Neurobiologia do Desenvolvimento, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho 373, CCS. Bloco G, Rio de Janeiro, 21941-902, Brazil
| |
Collapse
|
31
|
Elasticity-dependent response of malignant cells to viscous dissipation. Biomech Model Mechanobiol 2020; 20:145-154. [PMID: 32785801 PMCID: PMC7892690 DOI: 10.1007/s10237-020-01374-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/02/2020] [Accepted: 07/31/2020] [Indexed: 12/11/2022]
Abstract
The stiffness of the cellular environment controls malignant cell phenotype and proliferation. However, the effect of viscous dissipation on these parameters has not yet been investigated, in part due to the lack of in vitro cell substrates reproducing the mechanical properties of normal tissues and tumors. In this article, we use a newly reported viscoelastic polyacrylamide gel cell substrate, and we characterize the impact of viscous dissipation on three malignant cell lines: DU145 and PC3 derived from prostate and LN229 from brain. The spreading, motility and proliferation rates of these cells were analyzed on 1 kPa and 5 kPa elastic and viscoelastic gels. Surprisingly, the effect of substrate viscous dissipation on cell behavior depended on substrate stiffness for the three cell types tested. We conclude that viscoelasticity controls the spreading, proliferation and migration of malignant cells in vitro. These results highlight the critical role of viscous dissipation in the phenotype and proliferation of malignant cells, especially in stiff tumor environments.
Collapse
|
32
|
Xia J, Yuan Y, Wu H, Huang Y, Weitz DA. Decoupling the effects of nanopore size and surface roughness on the attachment, spreading and differentiation of bone marrow-derived stem cells. Biomaterials 2020; 248:120014. [PMID: 32276040 PMCID: PMC7262959 DOI: 10.1016/j.biomaterials.2020.120014] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/29/2019] [Revised: 03/24/2020] [Accepted: 03/27/2020] [Indexed: 12/25/2022]
Abstract
The nanopore size and roughness of nanoporous surface are two critical variables in determining stem cell fate, but little is known about the contribution from each cue individually. To address this gap, we use two-dimensional nanoporous membranes with controlled nanopore size and roughness to culture bone marrow-derived mesenchymal stem cells (BMSCs), and study their behaviors such as attachment, spreading and differentiation. We find that increasing the roughness of nanoporous surface has no noticeable effect on cell attachment, and only slightly decreases cell spreading areas and inhibits osteogenic differentiation. However, BMSCs cultured on membranes with larger nanopores have significantly fewer attached cells and larger spreading areas. Moreover, these cells cultured on larger nanopores undergo enhanced osteogenic differentiation by expressing more alkaline phosphatase, osteocalcin, osteopontin, and secreting more collagen type I. These results suggest that although both nanopore size and roughness can affect BMSCs, nanopore size plays a more significant role than roughness in controlling BMSC behavior.
Collapse
Affiliation(s)
- Jing Xia
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Yuan Yuan
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Huayin Wu
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Yuting Huang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - David A Weitz
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA; Department of Physics, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
33
|
Uddin Z, Fang T, Siao J, Tseng W. Wound Healing Attributes of Polyelectrolyte Multilayers Prepared with Multi‐
l
‐arginyl‐poly‐
l
‐aspartate Pairing with Hyaluronic Acid and γ‐Polyglutamic Acid. Macromol Biosci 2020; 20:e2000132. [DOI: 10.1002/mabi.202000132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/16/2020] [Revised: 05/26/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Zeeshan Uddin
- Department of Chemical EngineeringNational Taiwan University of Science and Technology No. 43, Sec. 4, Keelung Rd Taipei 106 Taiwan
| | - Tsuei‐Yun Fang
- Department of Food ScienceNational Taiwan Ocean University No. 2, Beining Rd Keelung 202 Taiwan
| | - Jyun‐Yin Siao
- Department of Chemical EngineeringNational Taiwan University of Science and Technology No. 43, Sec. 4, Keelung Rd Taipei 106 Taiwan
| | - Wen‐Chi Tseng
- Department of Chemical EngineeringNational Taiwan University of Science and Technology No. 43, Sec. 4, Keelung Rd Taipei 106 Taiwan
| |
Collapse
|
34
|
Bilodeau P, Jacobsen D, Law-Vinh D, Lee JM. Phosphatidylinositol 4-kinase III beta regulates cell shape, migration, and focal adhesion number. Mol Biol Cell 2020; 31:1904-1916. [PMID: 32583740 PMCID: PMC7525810 DOI: 10.1091/mbc.e19-11-0600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/11/2022] Open
Abstract
Cell shape is regulated by cell adhesion and cytoskeletal and membrane dynamics. Cell shape, adhesion, and motility have a complex relationship and understanding them is important in understanding developmental patterning and embryogenesis. Here we show that the lipid kinase phosphatidylinositol 4-kinase III beta (PI4KIIIβ) regulates cell shape, migration, and focal adhesion (FA) number. PI4KIIIβ generates phosphatidylinositol 4-phosphate (PI4P) from phosphatidylinositol and is highly expressed in a subset of human breast cancers. PI4KIIIβ and the PI4P it generates regulate a variety of cellular functions, ranging from control of Golgi structure, fly fertility, and Akt signaling. Here, we show that loss of PI4KIIIβ expression decreases cell migration and alters cell shape in NIH3T3 fibroblasts. The changes are accompanied by an increase in the number of FA in cells lacking PI4KIIIβ. Furthermore, we find that PI4P-containing vesicles move to the migratory leading edge during migration and that some of these vesicles tether to and fuse with FA. Fusion is associated with FA disassembly. This suggests a novel regulatory role for PI4KIIIβ and PI4P in cell adhesion and cell shape maintenance.
Collapse
Affiliation(s)
- Patricia Bilodeau
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daniel Jacobsen
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Denise Law-Vinh
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Jonathan M Lee
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
35
|
Yun YS, Kang EH, Ji S, Lee SB, Kim YO, Yun IS, Yeo JS. Quantitative Correlation of Nanotopography with Cell Spreading via Focal Adhesions Using Adipose-Derived Stem Cells. ACTA ACUST UNITED AC 2020; 4:e2000092. [PMID: 32500640 DOI: 10.1002/adbi.202000092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/29/2020] [Revised: 05/13/2020] [Indexed: 11/09/2022]
Abstract
Nanotopography mimicking extracellular environments reportedly impact cell morphological changes; however, elucidating this relationship has been challenging. To control cellular responses using nanostructures, in this study, the quantitative relationship between nanotopography and cell spreading mediated by focal adhesions (FAs) is demonstrated using adipose-derived stem cells (ASCs). The spreading of ASCs and area of FAs are analyzed for the distribution of filamentous actin and vinculin, respectively, using fluorescent images. FAs require a specific area for adhesion (herein defined as effective contact area [ECA]) to maintain cell attachment on nanopillar arrays. An ECA is the area of FAs supported by nanopillars, multiplying the area fraction (AF) of their top surface. Regarding the spreading of cells, the mean area of ASCs linearly decreases as the mean area of FAs increases. Because the area of FAs is inversely correlated to the AF of the nanopillar arrays, the spreading of cells can be quantitatively correlated with nanotopography. The results provide a conceptual framework for controlling cell behaviors to design artificial substrates for tissue-engineering applications.
Collapse
Affiliation(s)
- Young-Shik Yun
- School of Integrated Technology, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Korea.,Yonsei Institute of Convergence Technology, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Korea
| | - Eun-Hye Kang
- Department of Plastic and Reconstruction Surgery, College of Medicine, Yonsei University, 134, Sinchon-dong, Seodaemun-gu, Seoul, 03722, Korea
| | - Seungmuk Ji
- School of Integrated Technology, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Korea.,Yonsei Institute of Convergence Technology, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Korea
| | - Su-Bong Lee
- School of Integrated Technology, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Korea.,Yonsei Institute of Convergence Technology, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Korea
| | - Yong Oock Kim
- Department of Plastic and Reconstruction Surgery, College of Medicine, Yonsei University, 134, Sinchon-dong, Seodaemun-gu, Seoul, 03722, Korea
| | - In Sik Yun
- Department of Plastic and Reconstruction Surgery, College of Medicine, Yonsei University, 134, Sinchon-dong, Seodaemun-gu, Seoul, 03722, Korea
| | - Jong-Souk Yeo
- School of Integrated Technology, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Korea.,Yonsei Institute of Convergence Technology, Yonsei University, 85, Songdogwahak-ro, Yeonsu-gu, Incheon, 21983, Korea
| |
Collapse
|
36
|
Gerlach BD, Tubbesing K, Liao G, Rezey AC, Wang R, Barroso M, Tang DD. Phosphorylation of GMFγ by c-Abl Coordinates Lamellipodial and Focal Adhesion Dynamics to Regulate Airway Smooth Muscle Cell Migration. Am J Respir Cell Mol Biol 2020; 61:219-231. [PMID: 30811945 DOI: 10.1165/rcmb.2018-0352oc] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022] Open
Abstract
Airway smooth muscle cells require coordinated protrusion and focal adhesion dynamics to migrate properly. However, the signaling cascades that connect these two processes remain incompletely understood. Glia maturation factor (GMF)-γ has been implicated in inducing actin debranching and inhibiting nucleation. In this study, we discovered that GMFγ phosphorylation at Y104 regulates human airway smooth muscle cell migration. Using high-resolution microscopy coupled with three-dimensional object-based quantitative image analysis software, Imaris 9.2.0, phosphomimetic mutant, Y104D-GMFγ, was enriched at nascent adhesions along the leading edge where it recruited activated neural Wiskott-Aldrich syndrome protein (N-WASP; pY256) to promote actin-branch formation, which enhanced lamellipodial dynamics and limited the growth of focal adhesions. Unexpectedly, we found that nonphosphorylated mutant, Y104F-GMFγ, was enriched in growing adhesions where it promoted a linear branch organization and focal adhesion clustering, and recruited zyxin to increase maturation, thus inhibiting lamellipodial dynamics and cell migration. The localization of GMFγ between the leading edge and focal adhesions was dependent upon myosin activity. Furthermore, c-Abl tyrosine kinase regulated the GMFγ phosphorylation-dependent processes. Together, these results unveil the importance of GMFγ phosphorylation in coordinating lamellipodial and focal adhesion dynamics to regulate cell migration.
Collapse
Affiliation(s)
- Brennan D Gerlach
- Department of Molecular Cellular Physiology, Albany Medical College, Albany, New York
| | - Kate Tubbesing
- Department of Molecular Cellular Physiology, Albany Medical College, Albany, New York
| | - Guoning Liao
- Department of Molecular Cellular Physiology, Albany Medical College, Albany, New York
| | - Alyssa C Rezey
- Department of Molecular Cellular Physiology, Albany Medical College, Albany, New York
| | - Ruping Wang
- Department of Molecular Cellular Physiology, Albany Medical College, Albany, New York
| | - Margarida Barroso
- Department of Molecular Cellular Physiology, Albany Medical College, Albany, New York
| | - Dale D Tang
- Department of Molecular Cellular Physiology, Albany Medical College, Albany, New York
| |
Collapse
|
37
|
Girón-Pérez DA, Vadillo E, Schnoor M, Santos-Argumedo L. Myo1e modulates the recruitment of activated B cells to inguinal lymph nodes. J Cell Sci 2020; 133:jcs.235275. [PMID: 31964710 DOI: 10.1242/jcs.235275] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/12/2019] [Accepted: 01/06/2020] [Indexed: 12/31/2022] Open
Abstract
The inclusion of lymphocytes in high endothelial venules and their migration to the lymph nodes are critical steps in the immune response. Cell migration is regulated by the actin cytoskeleton and myosins. Myo1e is a long-tailed class I myosin and is highly expressed in B cells, which have not been studied in the context of cell migration. By using intravital microscopy in an in vivo model and performing in vitro experiments, we studied the relevance of Myo1e for the adhesion and inclusion of activated B cells in high endothelial venules. We observed reduced expression of integrins and F-actin in the membrane protrusions of B lymphocytes, which might be explained by deficiencies in vesicular trafficking. Interestingly, the lack of Myo1e reduced the phosphorylation of focal adhesion kinase (FAK; also known as PTK2), AKT (also known as AKT1) and RAC-1, disturbing the FAK-PI3K-RAC-1 signaling pathway. Taken together, our results indicate a critical role of Myo1e in the mechanism of B-cell adhesion and migration.
Collapse
Affiliation(s)
- Daniel A Girón-Pérez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, CP 07360, Mexico City, Mexico
| | - Eduardo Vadillo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, CP 07360, Mexico City, Mexico
| | - Michael Schnoor
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, CP 07360, Mexico City, Mexico
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, CP 07360, Mexico City, Mexico
| |
Collapse
|
38
|
Rizwan M, Yao Y, Gorbet MB, Tse J, Anderson DEJ, Hinds MT, Yim EKF. One-Pot Covalent Grafting of Gelatin on Poly(Vinyl Alcohol) Hydrogel to Enhance Endothelialization and Hemocompatibility for Synthetic Vascular Graft Applications. ACS APPLIED BIO MATERIALS 2020; 3:693-703. [PMID: 32656504 PMCID: PMC7351135 DOI: 10.1021/acsabm.9b01026] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/29/2022]
Abstract
Cardiovascular diseases remain the leading cause of death worldwide. Patency rates of clinically-utilized small diameter synthetic vascular grafts such as Dacron® and expanded polytetrafluoroethylene (ePTFE) to treat cardiovascular disease are inadequate due to lack of endothelialization. Sodium trimetaphosphate (STMP) crosslinked PVA could be potentially employed as blood-compatible small diameter vascular graft for the treatment of cardiovascular disease. However, PVA severely lacks cell adhesion properties, and the efforts to endothelialize STMP-PVA have been insufficient to produce a functioning endothelium. To this end, we developed a one-pot method to conjugate cell-adhesive protein via hydroxyl-to-amine coupling using carbonyldiimidazole by targeting residual hydroxyl groups on crosslinked STMP-PVA hydrogel. Primary human umbilical vascular endothelial cells (HUVECs) demonstrated significantly improved cells adhesion, viability and spreading on modified PVA. Cells formed a confluent endothelial monolayer, and expressed vinculin focal adhesions, cell-cell junction protein zonula occludens 1 (ZO1), and vascular endothelial cadherin (VE-Cadherin). Extensive characterization of the blood-compatibility was performed on modified PVA hydrogel by examining platelet activation, platelet microparticle formation, platelet CD61 and CD62P expression, and thrombin generation, which showed that the modified PVA was blood-compatible. Additionally, grafts were tested under whole, flowing blood without any anticoagulants in a non-human primate, arteriovenous shunt model. No differences were seen in platelet or fibrin accumulation between the modified-PVA, unmodified PVA or clinical, ePTFE controls. This study presents a significant step in the modification of PVA for the development of next generation in situ endothelialized synthetic vascular grafts.
Collapse
Affiliation(s)
- Muhammad Rizwan
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| | - Yuan Yao
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| | - Maud B. Gorbet
- Systems Design Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
- Centre for Bioengineering and Biotechnology, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| | - John Tse
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| | - Deirdre E. J. Anderson
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Evelyn K. F. Yim
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, Canada N2L 3G1
| |
Collapse
|
39
|
Guo W, Yu H, Zhang L, Chen X, Liu Y, Wang Y, Zhang Y. Effect of hyperoside on cervical cancer cells and transcriptome analysis of differentially expressed genes. Cancer Cell Int 2019; 19:235. [PMID: 31516392 PMCID: PMC6734331 DOI: 10.1186/s12935-019-0953-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/07/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
Background Hyperoside (Hy) is a plant-derived quercetin 3-d-galactoside that exhibits inhibitory activities on various tumor types. The objective of the current study was to explore Hy effects on cervical cancer cell proliferation, and to perform a transcriptome analysis of differentially expressed genes. Methods Cervical cancer HeLa and C-33A cells were cultured and the effect of Hy treatment was determined using the Cell Counting Kit-8 (CCK-8) assay. After calculating the IC50 of Hy in HeLa and C-33A cells, the more sensitive to Hy treatment cell type was selected for RNA-Seq. Differentially expressed genes (DEGs) were identified by comparing gene expression between the Hy and control groups. Candidate genes were determined through DEG analysis, protein interaction network (PPI) construction, PPI module analysis, transcription factor (TF) prediction, TF-target network construction, and survival analysis. Finally, the key candidate genes were verified by RT-qPCR and western blot. Results Hy inhibited HeLa and C33A cell proliferation in a dose- and time-dependent manner, as determined by the CCK-8 assay. Treatment of C-33A cells with 2 mM Hy was selected for the subsequent experiments. Compared with the control group, 754 upregulated and 509 downregulated genes were identified after RNA-Seq. After functional enrichment, 74 gene ontology biological processes and 43 Kyoto Encyclopedia of Genes and Genomes pathways were obtained. According to the protein interaction network (PPI), PPI module analysis, TF-target network construction, and survival analysis, the key genes MYC, CNKN1A, PAX2, TFRC, ACOX2, UNC5B, APBA1, PRKACA, PEAR1, COL12A1, CACNA1G, PEAR1, and CCNA2 were detected. RT-qPCR was performed on the key genes, and Western blot was used to verify C-MYC and TFRC. C-MYC and TFRC expressions were lower and higher than the corresponding values in the control group, respectively, in accordance with the results from the RNA-Seq analysis. Conclusion Hy inhibited HeLa and C-33A cell proliferation through C-MYC gene expression reduction in C-33A cells and TFRC regulation. The results of the current study provide a theoretical basis for Hy treatment of cervical cancer.
Collapse
Affiliation(s)
- Weikang Guo
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Hui Yu
- 2Department of Cardiopulmonary Function, Harbin Medical University Cancer Hospital, Harbin, 150081 Heilongjiang Province China
| | - Lu Zhang
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Xiuwei Chen
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yunduo Liu
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yaoxian Wang
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| | - Yunyan Zhang
- 1Department of Gynecology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081 Heilongjiang Province China
| |
Collapse
|
40
|
Abstract
The cytoskeleton of animal cells is one of the most complicated and functionally versatile structures, involved in processes such as endocytosis, cell division, intra-cellular transport, motility, force transmission, reaction to external forces, adhesion and preservation, and adaptation of cell shape. These functions are mediated by three classical cytoskeletal filament types, as follows: Actin, microtubules, and intermediate filaments. The named filaments form a network that is highly structured and dynamic, responding to external and internal cues with a quick reorganization that is orchestrated on the time scale of minutes and has to be tightly regulated. Especially in brain tumors, the cytoskeleton plays an important role in spreading and migration of tumor cells. As the cytoskeletal organization and regulation is complex and many-faceted, this review aims to summarize the findings about cytoskeletal filament types, including substructures formed by them, such as lamellipodia, stress fibers, and interactions between intermediate filaments, microtubules and actin. Additionally, crucial regulatory aspects of the cytoskeletal filaments and the formed substructures are discussed and integrated into the concepts of cell motility. Even though little is known about the impact of cytoskeletal alterations on the progress of glioma, a final point discussed will be the impact of established cytoskeletal alterations in the cellular behavior and invasion of glioma.
Collapse
|
41
|
Ibrahim M, Schoelermann J, Mustafa K, Cimpan MR. TiO
2
nanoparticles disrupt cell adhesion and the architecture of cytoskeletal networks of human osteoblast‐like cells in a size dependent manner. J Biomed Mater Res A 2018; 106:2582-2593. [DOI: 10.1002/jbm.a.36448] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/08/2017] [Revised: 04/07/2018] [Accepted: 04/25/2018] [Indexed: 12/17/2022]
Affiliation(s)
- Mohamed Ibrahim
- Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenBergen Norway
- Centre for International Health, Department of Global Public Health and Primary Care, Faculty of MedicineUniversity of BergenBergen Norway
| | - Julia Schoelermann
- Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenBergen Norway
- BerGenBio ASBergen Norway
| | - Kamal Mustafa
- Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenBergen Norway
| | - Mihaela R. Cimpan
- Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenBergen Norway
| |
Collapse
|
42
|
Andasari V, Lü D, Swat M, Feng S, Spill F, Chen L, Luo X, Zaman M, Long M. Computational model of wound healing: EGF secreted by fibroblasts promotes delayed re-epithelialization of epithelial keratinocytes. Integr Biol (Camb) 2018; 10:605-634. [PMID: 30206629 PMCID: PMC6571173 DOI: 10.1039/c8ib00048d] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022]
Abstract
It is widely agreed that keratinocyte migration plays a crucial role in wound re-epithelialization. Defects in this function contribute to wound reoccurrence causing significant clinical problems. Several in vitro studies have shown that the speed of migrating keratinocytes can be regulated by epidermal growth factor (EGF) which affects keratinocyte's integrin expression. The relationship between integrin expression (through cell-matrix adhesion) stimulated by EGF and keratinocyte migration speed is not linear since increased adhesion, due to increased integrin expression, has been experimentally shown to slow down cell migration due to the biphasic dependence of cell speed on adhesion. In our previous work we showed that keratinocytes that were co-cultured with EGF-enhanced fibroblasts formed an asymmetric migration pattern, where, the cumulative distances of keratinocytes migrating toward fibroblasts were smaller than those migrating away from fibroblasts. This asymmetric pattern is thought to be provoked by high EGF concentration secreted by fibroblasts. The EGF stimulates the expression of integrin receptors on the surface of keratinocytes migrating toward fibroblasts via paracrine signaling. In this paper, we present a computational model of keratinocyte migration that is controlled by EGF secreted by fibroblasts using the Cellular Potts Model (CPM). Our computational simulation results confirm the asymmetric pattern observed in experiments. These results provide a deeper insight into our understanding of the complexity of keratinocyte migration in the presence of growth factor gradients and may explain re-epithelialization failure in impaired wound healing.
Collapse
Affiliation(s)
- Vivi Andasari
- Boston University, Department of Biomedical Engineering, 44 Cummington Mall, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Three-dimensional (3D) cell culture systems have gained increasing interest not only for 3D migration studies but also for their use in drug screening, tissue engineering, and ex vivo modeling of metastatic behavior in the field of cancer biology and morphogenesis in the field of developmental biology. The goal of studying cells in a 3D context is to attempt to more faithfully recapitulate the physiological microenvironment of tissues, including mechanical and structural parameters that we envision will reveal more predictive data for development programs and disease states. In this review, we discuss the pros and cons of several well-characterized 3D cell culture systems for performing 3D migration studies. We discuss the intracellular and extracellular signaling mechanisms that govern cell migration. We also describe the mathematical models and relevant assumptions that can be used to describe 3D cell movement.
Collapse
Affiliation(s)
- Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences in Oncology Center, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, USA;, ,
| | - Daniele M. Gilkes
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences in Oncology Center, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, USA;, ,
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences in Oncology Center, Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, USA;, ,
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
44
|
Paim A, Braghirolli DI, Cardozo NSM, Pranke P, Tessaro IC. Human dental pulp stem cell adhesion and detachment in polycaprolactone electrospun scaffolds under direct perfusion. ACTA ACUST UNITED AC 2018; 51:e6754. [PMID: 29590258 PMCID: PMC5886556 DOI: 10.1590/1414-431x20186754] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/12/2017] [Accepted: 01/11/2018] [Indexed: 11/22/2022]
Abstract
Cell adhesion in three-dimensional scaffolds plays a key role in tissue development. However, stem cell behavior in electrospun scaffolds under perfusion is not fully understood. Thus, an investigation was made on the effect of flow rate and shear stress, adhesion time, and seeding density under direct perfusion in polycaprolactone electrospun scaffolds on human dental pulp stem cell detachment. Polycaprolactone scaffolds were electrospun using a solvent mixture of chloroform and methanol. The viable cell number was determined at each tested condition. Cell morphology was analyzed by confocal microscopy after various incubation times for static cell adhesion with a high seeding density. Scanning electron microscopy images were obtained before and after perfusion for the highest flow rate tested. The wall pore shear stress was calculated for all tested flow rates (0.005–3 mL/min). An inversely proportional relationship between adhesion time with cell detachment under perfusion was observed. Lower flow rates and lower seeding densities reduced the drag of cells by shear stress. However, there was an operational limit for the lowest flow rate that can be used without compromising cell viability, indicating that a flow rate of 0.05 mL/min might be more suitable for the tested cell culture in electrospun scaffolds under direct perfusion.
Collapse
Affiliation(s)
- A Paim
- Laboratório de Separação por Membranas, Departamento de Engenharia Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil.,Laboratório de Simulação, Departamento de Engenharia Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil.,Laboratório de Hematologia e Células-Tronco, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - D I Braghirolli
- Laboratório de Hematologia e Células-Tronco, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - N S M Cardozo
- Laboratório de Simulação, Departamento de Engenharia Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - P Pranke
- Laboratório de Hematologia e Células-Tronco, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil.,Instituto de Pesquisa com Células-Tronco, Porto Alegre, RS, Brasil
| | - I C Tessaro
- Laboratório de Separação por Membranas, Departamento de Engenharia Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| |
Collapse
|
45
|
Cleghorn WM, Bulus N, Kook S, Gurevich VV, Zent R, Gurevich EV. Non-visual arrestins regulate the focal adhesion formation via small GTPases RhoA and Rac1 independently of GPCRs. Cell Signal 2018; 42:259-269. [PMID: 29133163 PMCID: PMC5732042 DOI: 10.1016/j.cellsig.2017.11.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/17/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
Abstract
Arrestins recruit a variety of signaling proteins to active phosphorylated G protein-coupled receptors in the plasma membrane and to the cytoskeleton. Loss of arrestins leads to decreased cell migration, altered cell shape, and an increase in focal adhesions. Small GTPases of the Rho family are molecular switches that regulate actin cytoskeleton and affect a variety of dynamic cellular functions including cell migration and cell morphology. Here we show that non-visual arrestins differentially regulate RhoA and Rac1 activity to promote cell spreading via actin reorganization, and focal adhesion formation via two distinct mechanisms. Arrestins regulate these small GTPases independently of G-protein-coupled receptor activation.
Collapse
Affiliation(s)
- Whitney M Cleghorn
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States
| | - Nada Bulus
- Department of Medicine, Vanderbilt University, Nashville, TN 37232, United States
| | - Seunghyi Kook
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States
| | - Roy Zent
- Department of Medicine, Vanderbilt University, Nashville, TN 37232, United States; Department of Veterans Affairs Hospital, Nashville, TN, 37232, United States
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States.
| |
Collapse
|
46
|
Kim JK, Louhghalam A, Lee G, Schafer BW, Wirtz D, Kim DH. Nuclear lamin A/C harnesses the perinuclear apical actin cables to protect nuclear morphology. Nat Commun 2017; 8:2123. [PMID: 29242553 PMCID: PMC5730574 DOI: 10.1038/s41467-017-02217-5] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/21/2017] [Accepted: 11/14/2017] [Indexed: 11/17/2022] Open
Abstract
The distinct spatial architecture of the apical actin cables (or actin cap) facilitates rapid biophysical signaling between extracellular mechanical stimuli and intracellular responses, including nuclear shaping, cytoskeletal remodeling, and the mechanotransduction of external forces into biochemical signals. These functions are abrogated in lamin A/C-deficient mouse embryonic fibroblasts that recapitulate the defective nuclear organization of laminopathies, featuring disruption of the actin cap. However, how nuclear lamin A/C mediates the ability of the actin cap to regulate nuclear morphology remains unclear. Here, we show that lamin A/C expressing cells can form an actin cap to resist nuclear deformation in response to physiological mechanical stresses. This study reveals how the nuclear lamin A/C-mediated formation of the perinuclear apical actin cables protects the nuclear structural integrity from extracellular physical disturbances. Our findings highlight the role of the physical interactions between the cytoskeletal network and the nucleus in cellular mechanical homeostasis.
Collapse
Affiliation(s)
- Jeong-Ki Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Arghavan Louhghalam
- Department of Civil and Environmental Engineering, University of Massachusetts Dartmouth, Dartmouth, MA, 02747, USA
| | - Geonhui Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Benjamin W Schafer
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Civil Engineering, The John Hopkins University, Baltimore, MD, 21218, USA
| | - Denis Wirtz
- Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Chemical and Biomolecular Engineering, The John Hopkins University, Baltimore, MD, 21218, USA
- Johns Hopkins Physical Sciences-Oncology Center, The Johns Hopkins University, Baltimore, MD, 21218, USA
- Departments of Pathology and Oncology and Sydney Kimmel Comprehensive Cancer Center, The Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea.
| |
Collapse
|
47
|
Zhong DN, Luo YH, Mo WJ, Zhang X, Tan Z, Zhao N, Pang SM, Chen G, Rong MH, Tang W. High expression of long non‑coding HOTAIR correlated with hepatocarcinogenesis and metastasis. Mol Med Rep 2017; 17:1148-1156. [PMID: 29115524 DOI: 10.3892/mmr.2017.7999] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/25/2017] [Accepted: 08/14/2017] [Indexed: 11/06/2022] Open
Abstract
HOX transcript antisense RNA (HOTAIR), a newly discovered long noncoding RNA (lncRNA), has been reported to be a poor prognostic marker in many types of cancers. The current study attempted to investigate the biological roles and clinicopathlogical implications of HOTAIR in hepatocellular carcinoma (HCC), as well as understand the molecular mechanisms of HOTAIR in HCC progression. HOTAIR expression in 95 HCC patients with paired HCC tissues and adjacent non‑cancer tissues were investigated using quantitative reverse transcription‑polymerase chain reaction. The association between HOTAIR expression and clinicopathological features was assessed. The effects of HOTAIR were examined in vitro assays by silencing the lncRNA. Pathway analyses were performed to illustrate the biological functions of the HOTAIR and coexpression genes. The expression level of HOTAIR was observed significantly higher in the HCC tissue than the adjacent non‑tumor tissue. HOTAIR expression levels were significantly higher in tumor samples from patients with distant metastasis, advanced stage, portal vein tumor embolus, vasoinvasion, tumor capsular infiltration or positive nm23 expression than those from patients without these conditions, correspondingly. The silencing of HOTAIR in liver cancer cells induced the inhibition of cell proliferation and promotion of apoptosis. Several pathways such as extracellular matrix‑receptor interaction, focal adhesion, pathways in cancer were annotated with the HOTAIR and coexpression genes. In summary, the present analysis indicates that HOTAIR might be an oncogene in HCC. It functions though promoting tumor cell growth and inhibiting apoptosis. HOTAIR may potentially be involved in HCC metastatic progression by several pathways correlated to cell adhesion, and may be a therapeutic target in future.
Collapse
Affiliation(s)
- Da-Ni Zhong
- Department of Chemotherapy, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yi-Huan Luo
- Research Department, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Wei-Jia Mo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xin Zhang
- Research Department, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Zhong Tan
- Research Department, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Na Zhao
- Research Department, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Si-Min Pang
- Research Department, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Min-Hua Rong
- Research Department, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Wei Tang
- Department of Breast Surgery, Affiliated Cancer Hospital, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
48
|
Abstract
Rho-associated kinase (ROCK) activity drives cell migration via actomyosin contractility. During invasion, individual cancer cells can transition between 2 modes of migration, mesenchymal and amoeboid. Changes in ROCK activity can cause a switch between these migration phenotypes which are defined by distinct morphologies. However, recent studies have shown that the ROCK isoforms are not functionally redundant as previously thought. Therefore, it is unclear whether the ROCK isoforms play different roles in regulating migration phenotypes. Here, we found that ROCK1 and ROCK2 differentially regulate carcinoma cell morphology resulting in intermediate phenotypes that share some mesenchymal and amoeboid characteristics. These findings suggest that the ROCK isoforms play unique roles in the phenotypic plasticity of mesenchymal carcinoma cells which may have therapeutic implications.
Collapse
Affiliation(s)
- Rachel J Jerrell
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mitchell J Leih
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aron Parekh
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
49
|
Grigoriou E, Cantini M, Dalby MJ, Petersen A, Salmeron-Sanchez M. Cell migration on material-driven fibronectin microenvironments. Biomater Sci 2017; 5:1326-1333. [PMID: 28612879 PMCID: PMC5858633 DOI: 10.1039/c7bm00333a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/12/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022]
Abstract
Cell migration is a fundamental process involved in a wide range of biological phenomena. However, how the underlying mechanisms that control migration are orchestrated is not fully understood. In this work, we explore the migratory characteristics of human fibroblasts using different organisations of fibronectin (FN) triggered by two chemically similar surfaces, poly(ethyl acrylate) (PEA) and poly(methyl acrylate) (PMA); cell migration is mediated via an intermediate layer of fibronectin (FN). FN is organised into nanonetworks upon simple adsorption on PEA whereas a globular conformation is observed on PMA. We studied cell speed over the course of 24 h and the morphology of focal adhesions in terms of area and length. Additionally, we analysed the amount of cell-secreted FN as well as FN remodelling. Velocity of human fibroblasts was found to exhibit a biphasic behaviour on PEA, whereas it remained fairly constant on PMA. FA analysis revealed more mature focal adhesions on PEA over time contrary to smaller FAs found on PMA. Finally, human fibroblasts seemed to remodel adsorbed FN more on PMA than on PEA. Overall, these results indicate that the cell-protein-material interface affects cell migratory behaviour. Analysis of FAs together with FN secretion and remodelling were associated with differences in cell velocity providing insights into the factors that can modulate cell motility.
Collapse
Affiliation(s)
- E. Grigoriou
- Division of Biomedical Engineering , School of Engineering , University of Glasgow , Glasgow , UK .
| | - M. Cantini
- Division of Biomedical Engineering , School of Engineering , University of Glasgow , Glasgow , UK .
| | - M. J. Dalby
- Centre for Cell Engineering , University of Glasgow , UK
| | - A. Petersen
- Berlin Brandenburg Center for Regenerative Therapies , Charité-Universitätsmedizin Berlin , Berlin , Germany .
| | - M. Salmeron-Sanchez
- Division of Biomedical Engineering , School of Engineering , University of Glasgow , Glasgow , UK .
| |
Collapse
|
50
|
Integrin-Dependent Regulation of Small GTPases: Role in Cell Migration. J Indian Inst Sci 2017. [DOI: 10.1007/s41745-016-0010-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/20/2022]
|