1
|
Okoro J, Bulusu R, Frimpong E, Zhu X, Rogers S, Agyare E. A novel gemcitabine analog as a potential anticancer agent: synthesis and in-vitro evaluation against pancreatic cancer. Am J Cancer Res 2025; 15:684-704. [PMID: 40084373 PMCID: PMC11897620 DOI: 10.62347/kxsr8930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/28/2024] [Indexed: 03/16/2025] Open
Abstract
Gemcitabine (Gem) is approved for use in pancreatic cancer chemotherapy. However, Gem undergoes rapid metabolism in the blood, producing an inactive metabolite. Due to this rapid metabolism, the effective dose of Gem is high, thereby predisposing patients to severe adverse effects. This study aimed to improve Gem's metabolic and therapeutic stability by modifying the amine group (4-NH2) with hydroxylamine to form 4-N-hydroxylGem hydrochloride (GemAGY). Micro-elemental analysis and Nuclear Magnetic Resonance (NMR) were used to characterize GemAGY, and its anticancer activity was investigated against MiaPaCa-2, BxPC-3, and PANC-1 pancreatic cancer cell lines. The GemAGY metabolic stability was evaluated in human liver microsomal solution. In the 2D cytotoxicity assay, the IC50 values of GemAGY-treated MiaPaCa-2, PANC-1, and BxPC-3 cells were significantly lower when compared to GemHCl-treated cultures. More so, in 3D spheroid assay results, GemAGY IC50 values were found to be 9.5 ± 1.1 µM and 12.6 ± 1.0 µM when compared to GemHCl IC50 values of 24.1 ± 1.6 µM and 30.2 ± 1.8 µM in MiaPaCa-2 and PANC-1 cells, respectively. GemAGY was stable, with 60% remaining intact after 2 hours of digestion in microsomal enzymes, compared to GemHCl, which had less than 45% remaining intact after 30 minutes. GemAGY-treated MiaPaCa-2 and PANC-1 cells at 3.12 and 6.25 μM concentrations demonstrated a significantly reduced cell migration towards the wound area compared to the GemHCl-treated cultures at the same concentrations. Further, GemAGY-treated MiaPaCa-2 cells significantly increased the expression of p53 and BAX compared to GemHCl-treated cells. GemAGY demonstrated significant anticancer activity and improved metabolic stability compared to GemHCl and is most likely to have potential anticancer activity against pancreatic cancer.
Collapse
Affiliation(s)
- Joy Okoro
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| | - Raviteja Bulusu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| | - Esther Frimpong
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| | - Xue Zhu
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| | - Sherise Rogers
- Department of Medicine, Division of Hematology and Oncology, University of Florida College of MedicineGainesville, Florida, USA
| | - Edward Agyare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M UniversityTallahassee, Florida, USA
| |
Collapse
|
2
|
Niu D, Zhang Y, Chen J, Li D, He C, Liu H. Mechanobiology Platform Realized Using Photomechanical Mxene Nanocomposites: Bilayer Photoactuator Design and In Vitro Mechanical Forces Stimulation. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6869. [PMID: 36234210 PMCID: PMC9570783 DOI: 10.3390/ma15196869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/25/2022] [Accepted: 09/06/2022] [Indexed: 06/16/2023]
Abstract
Mechanotransduction is the process by which cells convert external forces and physical constraints into biochemical signals that control several aspects of cellular behavior. A number of approaches have been proposed to investigate the mechanisms of mechanotransduction; however, it remains a great challenge to develop a platform for dynamic multivariate mechanical stimulation of single cells and small colonies of cells. In this study, we combined polydimethylsiloxane (PDMS) and PDMS/Mxene nanoplatelets (MNPs) to construct a soft bilayer nanocomposite for extracellular mechanical stimulation. Fast backlash actuation of the bilayer as a result of near-infrared irradiation caused mechanical force stimulation of cells in a controllable manner. The excellent controllability of the light intensity and frequency allowed backlash bending acceleration and frequency to be manipulated. As gastric gland carcinoma cell line MKN-45 was the research subject, mechanical force loading conditions could trigger apoptosis of the cells in a stimulation duration time-dependent manner. Cell apoptotic rates were positively related to the duration time. In the case of 6 min mechanical force loading, apoptotic cell percentage rose to 34.46% from 5.5% of the control. This approach helps apply extracellular mechanical forces, even with predesigned loading cycles, and provides a solution to study cell mechanotransduction in complex force conditions. It is also a promising therapeutic technique for combining physical therapy and biomechanics.
Collapse
Affiliation(s)
- Dong Niu
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yanli Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, Medical College, Xizang Minzu University, Xianyang 712082, China
| | - Jinlan Chen
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Dachao Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Chunmeng He
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Hongzhong Liu
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- The Joint Key Laboratory of Graphene, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
3
|
van der Putten C, van den Broek D, Kurniawan NA. Myofibroblast transdifferentiation of keratocytes results in slower migration and lower sensitivity to mesoscale curvatures. Front Cell Dev Biol 2022; 10:930373. [PMID: 35938166 PMCID: PMC9355510 DOI: 10.3389/fcell.2022.930373] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/01/2022] [Indexed: 11/15/2022] Open
Abstract
Functional tissue repair after injury or disease is governed by the regenerative or fibrotic response by cells within the tissue. In the case of corneal damage, keratocytes are a key cell type that determine the outcome of the remodeling response by either adapting to a fibroblast or myofibroblast phenotype. Although a growing body of literature indicates that geometrical cues in the environment can influence Myo(fibroblast) phenotype, there is a lack of knowledge on whether and how differentiated keratocyte phenotype is affected by the curved tissue geometry in the cornea. To address this gap, in this study we characterized the phenotype of fibroblastic and transforming growth factor β (TGFβ)-induced myofibroblastic keratocytes and studied their migration behavior on curved culture substrates with varying curvatures. Immunofluorescence staining and quantification of cell morphological parameters showed that, generally, fibroblastic keratocytes were more likely to elongate, whereas myofibroblastic keratocytes expressed more pronounced α smooth muscle actin (α-SMA) and actin stress fibers as well as more mature focal adhesions. Interestingly, keratocyte adhesion on convex structures was weak and unstable, whereas they adhered normally on flat and concave structures. On concave cylinders, fibroblastic keratocytes migrated faster and with higher persistence along the longitudinal direction compared to myofibroblastic keratocytes. Moreover, this behavior became more pronounced on smaller cylinders (i.e., higher curvatures). Taken together, both keratocyte phenotypes can sense and respond to the sign and magnitude of substrate curvatures, however, myofibroblastic keratocytes exhibit weaker curvature sensing and slower migration on curved substrates compared to fibroblastic keratocytes. These findings provide fundamental insights into keratocyte phenotype after injury, but also exemplify the potential of tuning the physical cell environments in tissue engineering settings to steer towards a favorable regeneration response.
Collapse
Affiliation(s)
- Cas van der Putten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Daniëlle van den Broek
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
- *Correspondence: Nicholas A. Kurniawan,
| |
Collapse
|
4
|
Chandra A, Butler MT, Bear JE, Haugh JM. Modeling cell protrusion predicts how myosin II and actin turnover affect adhesion-based signaling. Biophys J 2022; 121:102-118. [PMID: 34861242 PMCID: PMC8758409 DOI: 10.1016/j.bpj.2021.11.2889] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/03/2021] [Accepted: 11/29/2021] [Indexed: 01/07/2023] Open
Abstract
Orchestration of cell migration is essential for development, tissue regeneration, and the immune response. This dynamic process integrates adhesion, signaling, and cytoskeletal subprocesses across spatial and temporal scales. In mesenchymal cells, adhesion complexes bound to extracellular matrix mediate both biochemical signal transduction and physical interaction with the F-actin cytoskeleton. Here, we present a mathematical model that offers insight into both aspects, considering spatiotemporal dynamics of nascent adhesions, active signaling molecules, mechanical clutching, actin treadmilling, and nonmuscle myosin II contractility. At the core of the model is a positive feedback loop, whereby adhesion-based signaling promotes generation of barbed ends at, and protrusion of, the cell's leading edge, which in turn promotes formation and stabilization of nascent adhesions. The model predicts a switch-like transition and optimality of membrane protrusion, determined by the balance of actin polymerization and retrograde flow, with respect to extracellular matrix density. The model, together with new experimental measurements, explains how protrusion can be modulated by mechanical effects (nonmuscle myosin II contractility and adhesive bond stiffness) and F-actin turnover.
Collapse
Affiliation(s)
- Ankit Chandra
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina
| | - Mitchell T Butler
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - James E Bear
- Department of Cell Biology and Physiology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina.
| |
Collapse
|
5
|
Dessalles CA, Babataheri A, Barakat AI. Pericyte mechanics and mechanobiology. J Cell Sci 2021; 134:134/6/jcs240226. [PMID: 33753399 DOI: 10.1242/jcs.240226] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Pericytes are mural cells of the microvasculature, recognized by their thin processes and protruding cell body. Pericytes wrap around endothelial cells and play a central role in regulating various endothelial functions, including angiogenesis and inflammation. They also serve as a vascular support and regulate blood flow by contraction. Prior reviews have examined pericyte biological functions and biochemical signaling pathways. In this Review, we focus on the role of mechanics and mechanobiology in regulating pericyte function. After an overview of the morphology and structure of pericytes, we describe their interactions with both the basement membrane and endothelial cells. We then turn our attention to biophysical considerations, and describe contractile forces generated by pericytes, mechanical forces exerted on pericytes, and pericyte responses to these forces. Finally, we discuss 2D and 3D engineered in vitro models for studying pericyte mechano-responsiveness and underscore the need for more evolved models that provide improved understanding of pericyte function and dysfunction.
Collapse
Affiliation(s)
- Claire A Dessalles
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120, Palaiseau, France
| | - Avin Babataheri
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120, Palaiseau, France
| | - Abdul I Barakat
- LadHyX, CNRS, Ecole polytechnique, Institut polytechnique de Paris, 91120, Palaiseau, France
| |
Collapse
|
6
|
Dolega ME, Monnier S, Brunel B, Joanny JF, Recho P, Cappello G. Extracellular matrix in multicellular aggregates acts as a pressure sensor controlling cell proliferation and motility. eLife 2021; 10:63258. [PMID: 33704063 PMCID: PMC8064752 DOI: 10.7554/elife.63258] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Imposed deformations play an important role in morphogenesis and tissue homeostasis, both in normal and pathological conditions. To perceive mechanical perturbations of different types and magnitudes, tissues need appropriate detectors, with a compliance that matches the perturbation amplitude. By comparing results of selective osmotic compressions of CT26 mouse cells within multicellular aggregates and global aggregate compressions, we show that global compressions have a strong impact on the aggregates growth and internal cell motility, while selective compressions of same magnitude have almost no effect. Both compressions alter the volume of individual cells in the same way over a shor-timescale, but, by draining the water out of the extracellular matrix, the global one imposes a residual compressive mechanical stress on the cells over a long-timescale, while the selective one does not. We conclude that the extracellular matrix is as a sensor that mechanically regulates cell proliferation and migration in a 3D environment.
Collapse
Affiliation(s)
- Monika E Dolega
- Université Grenoble Alpes, Laboratoire Interdisciplinaire de Physique, CNRS, Grenoble, France
| | - Sylvain Monnier
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, VILLEURBANNE, France
| | - Benjamin Brunel
- Université Grenoble Alpes, Laboratoire Interdisciplinaire de Physique, CNRS, Grenoble, France
| | | | - Pierre Recho
- Université Grenoble Alpes, Laboratoire Interdisciplinaire de Physique, CNRS, Grenoble, France
| | - Giovanni Cappello
- Université Grenoble Alpes, Laboratoire Interdisciplinaire de Physique, CNRS, Grenoble, France
| |
Collapse
|
7
|
Efthymiou G, Radwanska A, Grapa AI, Beghelli-de la Forest Divonne S, Grall D, Schaub S, Hattab M, Pisano S, Poet M, Pisani DF, Counillon L, Descombes X, Blanc-Féraud L, Van Obberghen-Schilling E. Fibronectin Extra Domains tune cellular responses and confer topographically distinct features to fibril networks. J Cell Sci 2021; 134:jcs.252957. [PMID: 33526715 DOI: 10.1242/jcs.252957] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 01/15/2021] [Indexed: 12/25/2022] Open
Abstract
Cellular fibronectin (FN; also known as FN1) variants harboring one or two alternatively spliced so-called extra domains (EDB and EDA) play a central bioregulatory role during development, repair processes and fibrosis. Yet, how the extra domains impact fibrillar assembly and function of the molecule remains unclear. Leveraging a unique biological toolset and image analysis pipeline for direct comparison of the variants, we demonstrate that the presence of one or both extra domains impacts FN assembly, function and physical properties of the matrix. When presented to FN-null fibroblasts, extra domain-containing variants differentially regulate pH homeostasis, survival and TGF-β signaling by tuning the magnitude of cellular responses, rather than triggering independent molecular switches. Numerical analyses of fiber topologies highlight significant differences in variant-specific structural features and provide a first step for the development of a generative model of FN networks to unravel assembly mechanisms and investigate the physical and functional versatility of extracellular matrix landscapes.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | - Agata Radwanska
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | - Anca-Ioana Grapa
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France.,Université Côte d'Azur, Inria, CNRS, i3S, Nice 06902, France
| | | | - Dominique Grall
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | | | - Maurice Hattab
- Université Côte d'Azur, CNRS, INSERM, iBV, Nice 06108, France
| | - Sabrina Pisano
- Université Côte d'Azur, Inserm, CNRS, IRCAN, Nice 06107, France
| | - Mallorie Poet
- Université Côte d'Azur, Inserm, CNRS, IRCAN, Nice 06107, France
| | | | | | | | | | | |
Collapse
|
8
|
FITC-Dextran Release from Cell-Embedded Fibrin Hydrogels. Biomolecules 2021; 11:biom11020337. [PMID: 33672379 PMCID: PMC7926394 DOI: 10.3390/biom11020337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Fibrin hydrogel is a central biological material in tissue engineering and drug delivery applications. As such, fibrin is typically combined with cells and biomolecules targeted to the regenerated tissue. Previous studies have analyzed the release of different molecules from fibrin hydrogels; however, the effect of embedded cells on the release profile has yet to be quantitatively explored. This study focused on the release of Fluorescein isothiocyanate (FITC)-dextran (FD) 250 kDa from fibrin hydrogels, populated with different concentrations of fibroblast or endothelial cells, during a 48-h observation period. The addition of cells to fibrin gels decreased the overall release by a small percentage (by 7-15% for fibroblasts and 6-8% for endothelial cells) relative to acellular gels. The release profile was shown to be modulated by various cellular activities, including gel degradation and physical obstruction to diffusion. Cell-generated forces and matrix deformation (i.e., densification and fiber alignment) were not found to significantly influence the release profiles. This knowledge is expected to improve fibrin integration in tissue engineering and drug delivery applications by enabling predictions and ways to modulate the release profiles of various biomolecules.
Collapse
|
9
|
Yadav S, Ta HT, Nguyen N. Mechanobiology in cardiology: Micro‐ and nanotechnologies to probe mechanosignaling. VIEW 2021. [DOI: 10.1002/viw.20200080] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Sharda Yadav
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
| | - Hang T. Ta
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
- School of Environment and Science Griffith University Nathan Queensland Australia
| | - Nam‐Trung Nguyen
- Queensland Micro‐ and Nanotechnology Centre Griffith University Nathan Queensland Australia
| |
Collapse
|
10
|
D'Urso M, Kurniawan NA. Mechanical and Physical Regulation of Fibroblast-Myofibroblast Transition: From Cellular Mechanoresponse to Tissue Pathology. Front Bioeng Biotechnol 2020; 8:609653. [PMID: 33425874 PMCID: PMC7793682 DOI: 10.3389/fbioe.2020.609653] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023] Open
Abstract
Fibroblasts are cells present throughout the human body that are primarily responsible for the production and maintenance of the extracellular matrix (ECM) within the tissues. They have the capability to modify the mechanical properties of the ECM within the tissue and transition into myofibroblasts, a cell type that is associated with the development of fibrotic tissue through an acute increase of cell density and protein deposition. This transition from fibroblast to myofibroblast-a well-known cellular hallmark of the pathological state of tissues-and the environmental stimuli that can induce this transition have received a lot of attention, for example in the contexts of asthma and cardiac fibrosis. Recent efforts in understanding how cells sense their physical environment at the micro- and nano-scales have ushered in a new appreciation that the substrates on which the cells adhere provide not only passive influence, but also active stimulus that can affect fibroblast activation. These studies suggest that mechanical interactions at the cell-substrate interface play a key role in regulating this phenotype transition by changing the mechanical and morphological properties of the cells. Here, we briefly summarize the reported chemical and physical cues regulating fibroblast phenotype. We then argue that a better understanding of how cells mechanically interact with the substrate (mechanosensing) and how this influences cell behaviors (mechanotransduction) using well-defined platforms that decouple the physical stimuli from the chemical ones can provide a powerful tool to control the balance between physiological tissue regeneration and pathological fibrotic response.
Collapse
Affiliation(s)
- Mirko D'Urso
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Nicholas A. Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| |
Collapse
|
11
|
Zhang Y, Huang Z, Dong S, Liu Z, Liu Y, Tang L, Cheng T, Zhou X. Evaluation of Cell's Passability in the ECM Network. Biophys J 2020; 119:1056-1064. [PMID: 32891186 DOI: 10.1016/j.bpj.2020.07.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/17/2020] [Accepted: 07/27/2020] [Indexed: 11/28/2022] Open
Abstract
The microstructure of the extracellular matrix (ECM) plays a key role in affecting cell migration, especially nonproteolytic migration. It is difficult, however, to measure some properties of the ECM, such as stiffness and the passability for cell migration. On the basis of a network model of collagen fiber in the ECM, which has been well applied to simulate mechanical behaviors such as the stress-strain relationship, damage, and failure, we proposed a series of methods to study the microstructural properties containing pore size and pore stiffness and to search for the possible migration paths for cells. Finally, with a given criterion, we quantitatively evaluated the passability of the ECM network for cell migration. The fiber network model with a microstructure and the analysis method presented in this study further our understanding of and ability to evaluate the properties of an ECM network.
Collapse
Affiliation(s)
- Yongrou Zhang
- Guangdong Key Laboratory of Modern Control Technology, Guangdong Institute of Intelligent Manufacturing, Guangzhou, Guangdong, China; School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, Guangdong, China
| | - Zetao Huang
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Shoubin Dong
- School of Computer Science and Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Zejia Liu
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, Guangdong, China.
| | - Yiping Liu
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, Guangdong, China
| | - Liqun Tang
- School of Civil Engineering and Transportation, South China University of Technology, Guangzhou, Guangdong, China; State Key Laboratory of Subtropical Building Science, South China University of Technology, Guangzhou, Guangdong, China
| | - Taobo Cheng
- Guangdong Key Laboratory of Modern Control Technology, Guangdong Institute of Intelligent Manufacturing, Guangzhou, Guangdong, China
| | - Xuefeng Zhou
- Guangdong Key Laboratory of Modern Control Technology, Guangdong Institute of Intelligent Manufacturing, Guangzhou, Guangdong, China.
| |
Collapse
|
12
|
Lin Z, Luo G, Du W, Kong T, Liu C, Liu Z. Recent Advances in Microfluidic Platforms Applied in Cancer Metastasis: Circulating Tumor Cells' (CTCs) Isolation and Tumor-On-A-Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1903899. [PMID: 31747120 DOI: 10.1002/smll.201903899] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/13/2019] [Indexed: 05/03/2023]
Abstract
Cancer remains the leading cause of death worldwide despite the enormous efforts that are made in the development of cancer biology and anticancer therapeutic treatment. Furthermore, recent studies in oncology have focused on the complex cancer metastatic process as metastatic disease contributes to more than 90% of tumor-related death. In the metastatic process, isolation and analysis of circulating tumor cells (CTCs) play a vital role in diagnosis and prognosis of cancer patients at an early stage. To obtain relevant information on cancer metastasis and progression from CTCs, reliable approaches are required for CTC detection and isolation. Additionally, experimental platforms mimicking the tumor microenvironment in vitro give a better understanding of the metastatic microenvironment and antimetastatic drugs' screening. With the advancement of microfabrication and rapid prototyping, microfluidic techniques are now increasingly being exploited to study cancer metastasis as they allow precise control of fluids in small volume and rapid sample processing at relatively low cost and with high sensitivity. Recent advancements in microfluidic platforms utilized in various methods for CTCs' isolation and tumor models recapitulating the metastatic microenvironment (tumor-on-a-chip) are comprehensively reviewed. Future perspectives on microfluidics for cancer metastasis are proposed.
Collapse
Affiliation(s)
- Zhengjie Lin
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Guanyi Luo
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Weixiang Du
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Tiantian Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Changkun Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Zhou Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
13
|
Werner M, Kurniawan NA, Bouten CVC. Cellular Geometry Sensing at Different Length Scales and its Implications for Scaffold Design. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E963. [PMID: 32098110 PMCID: PMC7078773 DOI: 10.3390/ma13040963] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/22/2022]
Abstract
Geometrical cues provided by the intrinsic architecture of tissues and implanted biomaterials have a high relevance in controlling cellular behavior. Knowledge of how cells sense and subsequently respond to complex geometrical cues of various sizes and origins is needed to understand the role of the architecture of the extracellular environment as a cell-instructive parameter. This is of particular interest in the field of tissue engineering, where the success of scaffold-guided tissue regeneration largely depends on the formation of new tissue in a native-like organization in order to ensure proper tissue function. A well-considered internal scaffold design (i.e., the inner architecture of the porous structure) can largely contribute to the desired cell and tissue organization. Advances in scaffold production techniques for tissue engineering purposes in the last years have provided the possibility to accurately create scaffolds with defined macroscale external and microscale internal architectures. Using the knowledge of how cells sense geometrical cues of different size ranges can drive the rational design of scaffolds that control cellular and tissue architecture. This concise review addresses the recently gained knowledge of the sensory mechanisms of cells towards geometrical cues of different sizes (from the nanometer to millimeter scale) and points out how this insight can contribute to informed architectural scaffold designs.
Collapse
Affiliation(s)
- Maike Werner
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Nicholas A. Kurniawan
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| | - Carlijn V. C. Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AP Eindhoven, The Netherlands; (M.W.); (C.V.C.B.)
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AZ Eindhoven, The Netherlands
| |
Collapse
|
14
|
Rosalem GS, Las Casas EB, Lima TP, González-Torres LA. A mechanobiological model to study upstream cell migration guided by tensotaxis. Biomech Model Mechanobiol 2020; 19:1537-1549. [PMID: 32006123 DOI: 10.1007/s10237-020-01289-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 01/11/2020] [Indexed: 01/06/2023]
Abstract
Cell migration is a process of crucial importance for the human body. It is responsible for important processes such as wound healing and tumor metastasis. Migration may occur in response to stimuli of chemical, physical and mechanical nature occurring in the cellular microenvironment. The interstitial flow (IF) can generate mechanical stimuli in cells that influence the cell behavior and interactions of the cells with the extracellular matrix (ECM). One of the phenomena is upstream migration, which is observed in some tumors. In this work, we present a new approach to study the adherent cell migration in a porous medium using a mechanobiological model, attempting to understand if upstream migration can be generated exclusively by mechanical factors. The influence of IF on the behavior of cells and the extracellular matrix was considered. The model is based on a system of coupled nonlinear differential equations solved by the finite element method. Several simulations were performed to study the upstream cell migration and evaluate the effects of pressure, permeability, ECM stiffness and cellular concentration variations on the cell velocity. The results indicated that upstream migration can occur in the presence of mechanical stimuli generated by IF and that the tested parameters have a direct influence on the cellular velocity, especially the pressure and the permeability.
Collapse
Affiliation(s)
- Gabriel Santos Rosalem
- Department of Mechanical Engineering, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Thiago Parente Lima
- Institute of Science and Technology, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | | |
Collapse
|
15
|
Kurniawan NA. The ins and outs of engineering functional tissues and organs: evaluating the in-vitro and in-situ processes. Curr Opin Organ Transplant 2019; 24:590-597. [PMID: 31389812 PMCID: PMC6749960 DOI: 10.1097/mot.0000000000000690] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW For many disorders that result in loss of organ function, the only curative treatment is organ transplantation. However, this approach is severely limited by the shortage of donor organs. Tissue engineering has emerged as an alternative solution to this issue. This review discusses the concept of tissue engineering from a technical viewpoint and summarizes the state of the art as well as the current shortcomings, with the aim of identifying the key lessons that we can learn to further advance the engineering of functional tissues and organs. RECENT FINDINGS A plethora of tissue-engineering strategies have been recently developed. Notably, these strategies put different emphases on the in-vitro and in-situ processes (i.e. preimplantation and postimplantation) that take place during tissue formation. Biophysical and biomechanical interactions between the cells and the scaffold/biomaterial play a crucial role in all steps and have started to be exploited to steer tissue regeneration. SUMMARY Recent works have demonstrated the need to better understand the in-vitro and in-situ processes during tissue formation, in order to regenerate complex, functional organs with desired cellular organization and tissue architecture. A concerted effort from both fundamental and tissue-specific research has the potential to accelerate progress in the field.
Collapse
Affiliation(s)
- Nicholas A. Kurniawan
- Department of Biomedical Engineering
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
16
|
Werner M, Petersen A, Kurniawan NA, Bouten CVC. Cell-Perceived Substrate Curvature Dynamically Coordinates the Direction, Speed, and Persistence of Stromal Cell Migration. ACTA ACUST UNITED AC 2019; 3:e1900080. [PMID: 32648723 DOI: 10.1002/adbi.201900080] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/16/2019] [Indexed: 01/02/2023]
Abstract
Adherent cells residing within tissues or biomaterials are presented with 3D geometrical cues from their environment, often in the form of local surface curvatures. While there is growing evidence that cellular decision-making is influenced by substrate curvature, the effect of physiologically relevant, cell-scale anisotropic curvatures remains poorly understood. This study systematically explores the migration behavior of human bone marrow stromal cells (hBMSCs) on a library of anisotropic curved structures. Analysis of cell trajectories reveals that, on convex cylindrical structures, hBMSC migration speed and persistence are strongly governed by the cellular orientation on the curved structure, while migration on concave cylindrical structures is characterized by fast but non-aligned and non-persistent migration. Concurrent presentation of concave and convex substrates on toroidal structures induces migration in the direction where hBMSCs can most effectively avoid cell bending. These distinct migration behaviors are found to be universally explained by the cell-perceived substrate curvature, which on anisotropic curved structures is dependent on both the temporally varying cell orientation and the 3D cellular morphology. This work demonstrates that cell migration is dynamically guided by the perceived curvature of the underlying substrate, providing an important biomaterial design parameter for instructing cell migration in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Maike Werner
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AJ, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| | - Ansgar Petersen
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, D-13353 , Berlin, Germany
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AJ, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AJ, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, 5600 MB, Eindhoven, The Netherlands
| |
Collapse
|
17
|
Spatarelu CP, Zhang H, Trung Nguyen D, Han X, Liu R, Guo Q, Notbohm J, Fan J, Liu L, Chen Z. Biomechanics of Collective Cell Migration in Cancer Progression: Experimental and Computational Methods. ACS Biomater Sci Eng 2019; 5:3766-3787. [PMID: 32953985 PMCID: PMC7500334 DOI: 10.1021/acsbiomaterials.8b01428] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cell migration is essential for regulating many biological processes in physiological or pathological conditions, including embryonic development and cancer invasion. In vitro and in silico studies suggest that collective cell migration is associated with some biomechanical particularities such as restructuring of extracellular matrix (ECM), stress and force distribution profiles, and reorganization of the cytoskeleton. Therefore, the phenomenon could be understood by an in-depth study of cells' behavior determinants, including but not limited to mechanical cues from the environment and from fellow "travelers". This review article aims to cover the recent development of experimental and computational methods for studying the biomechanics of collective cell migration during cancer progression and invasion. We also summarized the tested hypotheses regarding the mechanism underlying collective cell migration enabled by these methods. Together, the paper enables a broad overview on the methods and tools currently available to unravel the biophysical mechanisms pertinent to cell collective migration as well as providing perspectives on future development toward eventually deciphering the key mechanisms behind the most lethal feature of cancer.
Collapse
Affiliation(s)
| | - Hao Zhang
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Dung Trung Nguyen
- Department of Engineering and Computer Science, Seattle Pacific University, Seattle, Washington 98119,
United States
| | - Xinyue Han
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Ruchuan Liu
- College of Physics, Chongqing University, Chongqing 400032, China
| | - Qiaohang Guo
- School of Materials Science and Engineering, Fujian University of Technology, Fuzhou 350014,
China
| | - Jacob Notbohm
- Department of Engineering Physics, University of Wisconsin—Madison, Madison, Wisconsin 53706,
United States
| | - Jing Fan
- Department of Mechanical Engineering, City College of City University of New York, New York 10031, United
States
| | - Liyu Liu
- College of Physics, Chongqing University, Chongqing 400032, China
| | - Zi Chen
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| |
Collapse
|
18
|
Biophysical properties of cells for cancer diagnosis. J Biomech 2019; 86:1-7. [PMID: 30803699 DOI: 10.1016/j.jbiomech.2019.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/23/2018] [Accepted: 02/09/2019] [Indexed: 02/06/2023]
Abstract
Biophysical properties associated with the microenvironment of a tumor has been recognized as an important modulator for cell behaviour and function. Particularly, tissue rigidity is important during tumor carcinogenesis as it affects the tumor's ability to metastasis. Multiple downstream pathways are affected with a difference in rigidity of the extracellular matrix. The insight into tumor mechanosignalling represents a promising field that may lead to novel approaches for cancer diagnostics. Measurement of rigidity of the extracellular matrix or the tissue is a potential diagnostics approach for cancer detection. Altered extracellular matrix states persist for a long period of time and have lower heterogeneity compared to protein or genetic markers, therefore are more reliable as biomarkers. On the other hand, measurement of different kinase associated proteins or transcripts provide an early insight into potential transition of cells towards metastasis. Co-localization of transcriptional factors like YAP/TAZ provide an insight to determine if the cells are undergoing metastatic changes. This review explains the unique biophysical properties of the tumor microenvironment that present the potential targets for the diagnosis of cancer.
Collapse
|
19
|
Yeoman BM, Katira P. A stochastic algorithm for accurately predicting path persistence of cells migrating in 3D matrix environments. PLoS One 2018; 13:e0207216. [PMID: 30440015 PMCID: PMC6237354 DOI: 10.1371/journal.pone.0207216] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 10/26/2018] [Indexed: 01/07/2023] Open
Abstract
Cell mobility plays a critical role in immune response, wound healing, and the rate of cancer metastasis and tumor progression. Mobility within a three-dimensional (3D) matrix environment can be characterized by the average velocity of cell migration and the persistence length of the path it follows. Computational models that aim to predict cell migration within such 3D environments need to be able predict both of these properties as a function of the various cellular and extra-cellular factors that influence the migration process. A large number of models have been developed to predict the velocity of cell migration driven by cellular protrusions in 3D environments. However, prediction of the persistence of a cell's path is a more tedious matter, as it requires simulating cells for a long time while they migrate through the model extra-cellular matrix (ECM). This can be a computationally expensive process, and only recently have there been attempts to quantify cell persistence as a function of key cellular or matrix properties. Here, we propose a new stochastic algorithm that can simulate and analyze 3D cell migration occurring over days with a computation time of minutes, opening new possibilities of testing and predicting long-term cell migration behavior as a function of a large variety of cell and matrix properties. In this model, the matrix elements are generated as needed and stochastically based on the biophysical and biochemical properties of the ECM the cell migrates through. This approach significantly reduces the computational resources required to track and calculate cell matrix interactions. Using this algorithm, we predict the effect of various cellular and matrix properties such as cell polarity, cell mechanoactivity, matrix fiber density, matrix stiffness, fiber alignment, and fiber binding site density on path persistence of cellular migration and the mean squared displacement of cells over long periods of time.
Collapse
Affiliation(s)
- Benjamin Michael Yeoman
- Mechanical Engineering Department, San Diego State University, San Diego, CA, United States of America
- Department of Bioengineering, University of California San Diego, San Diego, CA, United States of America
| | - Parag Katira
- Mechanical Engineering Department, San Diego State University, San Diego, CA, United States of America
- Computational Science Research Center, San Diego State University, San Diego, CA, United States of America
| |
Collapse
|
20
|
Sopher RS, Tokash H, Natan S, Sharabi M, Shelah O, Tchaicheeyan O, Lesman A. Nonlinear Elasticity of the ECM Fibers Facilitates Efficient Intercellular Communication. Biophys J 2018; 115:1357-1370. [PMID: 30217380 DOI: 10.1016/j.bpj.2018.07.036] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 07/09/2018] [Accepted: 07/25/2018] [Indexed: 11/18/2022] Open
Abstract
Biological cells embedded in fibrous matrices have been observed to form intercellular bands of dense and aligned fibers through which they mechanically interact over long distances. Such matrix-mediated cellular interactions have been shown to regulate various biological processes. This study aimed to explore the effects of elastic nonlinearity of the fibers contained in the extracellular matrix (ECM) on the transmission of mechanical loads between contracting cells. Based on our biological experiments, we developed a finite-element model of two contracting cells embedded within a fibrous network. The individual fibers were modeled as showing linear elasticity, compression microbuckling, tension stiffening, or both of the latter two. Fiber compression buckling resulted in smaller loads in the ECM, which were primarily directed toward the neighboring cell. These loads decreased with increasing cell-to-cell distance; when cells were >9 cell diameters apart, no such intercellular interaction was observed. Tension stiffening further contributed to directing the loads toward the neighboring cell, though to a smaller extent. The contraction of two neighboring cells resulted in mutual attraction forces, which were considerably increased by tension stiffening and decayed with increasing cell-to-cell distances. Nonlinear elasticity contributed also to the onset of force polarity on the cell boundaries, manifested by larger contractile forces pointing toward the neighboring cell. The density and alignment of the fibers within the intercellular band were greater when fibers buckled under compression, with tension stiffening further contributing to this structural remodeling. Although previous studies have established the role of the ECM nonlinear mechanical behavior in increasing the range of force transmission, our model demonstrates the contribution of nonlinear elasticity of biological gels to directional and efficient mechanical signal transfer between distant cells, and rehighlights the importance of using fibrous gels in experimental settings for facilitating intercellular communication. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Ran S Sopher
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Hanan Tokash
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Sari Natan
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Mirit Sharabi
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ortal Shelah
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Oren Tchaicheeyan
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ayelet Lesman
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
21
|
Werner M, Kurniawan NA, Korus G, Bouten CVC, Petersen A. Mesoscale substrate curvature overrules nanoscale contact guidance to direct bone marrow stromal cell migration. J R Soc Interface 2018; 15:20180162. [PMID: 30089684 PMCID: PMC6127159 DOI: 10.1098/rsif.2018.0162] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 07/13/2018] [Indexed: 12/19/2022] Open
Abstract
The intrinsic architecture of biological tissues and of implanted biomaterials provides cells with large-scale geometrical cues. To understand how cells are able to sense and respond to complex structural environments, a deeper insight into the cellular response to multi-scale and conflicting geometrical cues is needed. In this study, we subjected human bone marrow stromal cells (hBMSCs) to mesoscale cylindrical surfaces (diameter 250-5000 µm) and nanoscale collagen fibrils (diameter 100-200 nm) that were aligned perpendicular to the cylinder axis. On flat surfaces and at low substrate curvatures (cylinder diameter d > 1000 µm), cell alignment and migration were governed by the nanoscale collagen fibrils, consistent with the contact guidance effect. With increasing surface curvature (decreasing cylinder diameter, d < 1000 µm), cells increasingly aligned and migrated along the cylinder axis, i.e. the direction of zero curvature. An increase in phosphorylated myosin light chain levels was observed with increasing substrate curvature, suggesting a link between substrate-induced cell bending and the F-actin-myosin machinery. Taken together, this work demonstrates that geometrical cues of up to 10× cell size can play a dominant role in directing hBMSC alignment and migration and that the effect of nanoscale contact guidance can even be overruled by mesoscale curvature guidance.
Collapse
Affiliation(s)
- Maike Werner
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, The Netherlands
| | - Nicholas A Kurniawan
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, The Netherlands
| | - Gabriela Korus
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Germany
| | - Carlijn V C Bouten
- Soft Tissue Engineering and Mechanobiology, Department of Biomedical Engineering, Eindhoven University of Technology, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, The Netherlands
| | - Ansgar Petersen
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Germany
| |
Collapse
|
22
|
Pakshir P, Hinz B. The big five in fibrosis: Macrophages, myofibroblasts, matrix, mechanics, and miscommunication. Matrix Biol 2018; 68-69:81-93. [DOI: 10.1016/j.matbio.2018.01.019] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 02/07/2023]
|
23
|
Affiliation(s)
- Parthiv Kant Chaudhuri
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Level 9, Singapore 117411, Singapore
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| | - Boon Chuan Low
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Level 9, Singapore 117411, Singapore
- Cell Signaling and Developmental Biology Laboratory, Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
- University Scholars Programme, National University of Singapore, Singapore 138593, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Level 9, Singapore 117411, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
- Biomedical Institute for Global Health Research and Technology (BIGHEART), National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
24
|
Hong X, Annamalai RT, Kemerer TS, Deng CX, Stegemann JP. Multimode ultrasound viscoelastography for three-dimensional interrogation of microscale mechanical properties in heterogeneous biomaterials. Biomaterials 2018; 178:11-22. [PMID: 29902533 DOI: 10.1016/j.biomaterials.2018.05.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 05/02/2018] [Accepted: 05/31/2018] [Indexed: 02/07/2023]
Abstract
Both static and time-dependent mechanical factors can have a profound impact on cell and tissue function, but it is challenging to measure the mechanical properties of soft materials at the scale which cells sense. Multimode ultrasound viscoelastography (MUVE) uses focused ultrasound pulses to both generate and image deformations within soft hydrogels non-invasively, at sub-millimeter resolution, and in 3D. The deformation and strain over time data are used to extract quantitative parameters that describe both the elastic and viscoelastic properties of the material. MUVE was used in creep mode to characterize the viscoelastic properties of 3D agarose, collagen, and fibrin hydrogels. Quantitative comparisons were made by extracting characteristic viscoelastic parameters using Burger's lumped parameter constitutive model. Spatial resolution of the MUVE technique was found to be approximately 200 μm, while detection sensitivity, defined as the capability to differentiate between materials based on mechanical property differences, was approximately 0.2 kPa using agarose hydrogels. MUVE was superior to nanoindentation and shear rheometry in generating consistent microscale measurements of viscoelastic behavior in soft materials. These results demonstrate that MUVE is a rapid, quantitative, and accurate method to measure the viscoelastic mechanical properties of soft 3D hydrogels at the microscale, and is a promising technique to study the development of native and engineered tissues over time.
Collapse
Affiliation(s)
- Xiaowei Hong
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, MI, MI 48109, USA
| | - Ramkumar T Annamalai
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, MI, MI 48109, USA
| | - Tyler S Kemerer
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, MI, MI 48109, USA
| | - Cheri X Deng
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, MI, MI 48109, USA.
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, MI, MI 48109, USA.
| |
Collapse
|
25
|
An automated quantitative analysis of cell, nucleus and focal adhesion morphology. PLoS One 2018; 13:e0195201. [PMID: 29601604 PMCID: PMC5877879 DOI: 10.1371/journal.pone.0195201] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 03/19/2018] [Indexed: 11/19/2022] Open
Abstract
Adherent cells sense the physical properties of their environment via focal adhesions. Improved understanding of how cells sense and response to their physical surroundings is aided by quantitative evaluation of focal adhesion size, number, orientation, and distribution in conjunction with the morphology of single cells and the corresponding nuclei. We developed a fast, user-friendly and automated image analysis algorithm capable of capturing and characterizing these individual components with a high level of accuracy. We demonstrate the robustness and applicability of the algorithm by quantifying morphological changes in response to a variety of environmental changes as well as manipulations of cellular components of mechanotransductions. Finally, as a proof-of-concept we use our algorithm to quantify the effect of Rho-associated kinase inhibitor Y-27632 on focal adhesion maturation. We show that a decrease in cell contractility leads to a decrease in focal adhesion size and aspect ratio.
Collapse
|
26
|
Miyoshi H, Suzuki K, Ju J, Ko JS, Adachi T, Yamagata Y. A Perturbation Analysis to Understand the Mechanism How Migrating Cells Sense and Respond to a Topography in the Extracellular Environment. ANAL SCI 2018; 32:1207-1211. [PMID: 27829627 DOI: 10.2116/analsci.32.1207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Migrating cells in vivo monitor the physiological state of an organism by integrating the physical as well as chemical cues in the extracellular microenvironment, and alter the migration mode, in order to achieve their unique function. The clarification of the mechanism focusing on the topographical cues is important for basic biological research, and for biomedical engineering specifically to establish the design concept of tissue engineering scaffolds. The aim of this study is to understand how cells sense and respond to the complex topographical cues in vivo by exploring in vitro analyses to complex in vivo situations in order to simplify the issue. Since the intracellular mechanical events at subcellular scales and the way of the coordination of these events are supposed to change in the migrating cells, a key to success of the analysis is a mechanical point of view with a particular focus of the subcellular mechanical events. We designed an experimental platform to explore the mechanical requirements in a migrating fibroma cell responding to micro-grooves. The micro-grooved structure is a model of gap structures, typically seen in the microenvironments in vivo. In our experiment, the contributions of actomyosin force generation can be spatially divided and analyzed in the cell center and peripheral regions. The analysis specified that rapid leading edge protrusion, and the cell body translocation coordinated with the leading edge protrusion are required for the turning response at a micro-groove.
Collapse
Affiliation(s)
- Hiromi Miyoshi
- Pathophysiological and Health Science Team, RIKEN Center for Life Science Technologies
| | | | | | | | | | | |
Collapse
|
27
|
ADAMTS-1 disrupts HGF/c-MET signaling and HGF-stimulated cellular processes in fibrosarcoma. Exp Cell Res 2018; 363:271-282. [PMID: 29355494 DOI: 10.1016/j.yexcr.2018.01.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/21/2017] [Accepted: 01/12/2018] [Indexed: 01/10/2023]
Abstract
Extracellular matrix (ECM) serves as a reservoir for biologically active factors, such as growth factors and proteases that influence the tumor cell behavior. ADAMTS-1 (a disintegrin and metalloprotease with thrombospondin motifs) is a secreted protease that has the ability to modify the ECM during physiological and pathological processes. Here, we analyzed the role played by ADAMTS-1 regulating HGF and TGF-β1 activities in the high-grade fibrosarcoma cell line (HT1080). We generated HT1080 and HEK293T cells overexpressing ADAMTS-1. HT1080 cells overexpressing ADAMTS-1 (HT1080-MPA) exhibited a significant decrease in cell proliferation and migration velocity, both in presence of HGF. We obtained similar results with ADAMTS-1-enriched conditioned medium from other cell type. However, ADAMTS-1 overexpression failed to affect TGF-β1 activity associated with HT1080 cell proliferation and migration velocity. Immunoblotting showed that ADAMTS-1 overexpression disturbs c-Met activation upon HGF stimulation. Downstream ERK1/2 and FAK signaling pathways are also influenced by this protease. Additionally, ADAMTS-1 decreased the size of the fibrosarcospheres, both under normal conditions and in the presence of HGF. Likewise, in presence of HGF, ADAMTS-1 overexpression in HT1080 disrupted microtumors formation in vivo. These microtumors, including individual cells, presented characteristics of non-invasive lesions (rounded morphology). Our results suggest that ADAMTS-1 is involved in regulating HGF-related functions on fibrosarcoma cells. This protease may then represent an endogenous mechanism in controlling the bioavailability of different growth factors that have a direct influence on tumor cell behavior.
Collapse
|
28
|
Zhao ZL, Liu ZY, Du J, Xu GK, Feng XQ. A Dynamic Biochemomechanical Model of Geometry-Confined Cell Spreading. Biophys J 2017; 112:2377-2386. [PMID: 28591610 DOI: 10.1016/j.bpj.2017.04.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/11/2017] [Accepted: 04/13/2017] [Indexed: 01/09/2023] Open
Abstract
Cell spreading is involved in many physiological and pathological processes. The spreading behavior of a cell significantly depends on its microenvironment, but the biochemomechanical mechanisms of geometry-confined cell spreading remain unclear. A dynamic model is here established to investigate the spreading of cells confined in a finite region with different geometries, e.g., rectangle, ellipse, triangle, and L-shape. This model incorporates both biophysical and biochemical mechanisms, including actin polymerization, integrin-mediated binding, plasma viscoelasticity, and the elasticity of membranes and microtubules. We simulate the dynamic configurational evolution of a cell under different geometric microenvironments, including the angular distribution of microtubule forces and the deformation of the nucleus. The results indicate that the positioning of the cell-division plane is affected by its boundary confinement: a cell divides in a plane perpendicular to its minimal principal axis of inertia of area. In addition, the effects of such physical factors as the adhesive bond density, membrane tension, and microtubule number are examined on the cell spreading dynamics. The theoretical predictions show a good agreement with relevant experimental results. This work sheds light on the geometry-confined spreading dynamics of cells and holds potential applications in regulating cell division and designing cell-based sensors.
Collapse
Affiliation(s)
- Zi-Long Zhao
- AML, Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, China; Center for Nano and Micro Mechanics, Tsinghua University, Beijing, China
| | - Zong-Yuan Liu
- AML, Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, China; Center for Nano and Micro Mechanics, Tsinghua University, Beijing, China
| | - Jing Du
- AML, Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, China
| | - Guang-Kui Xu
- International Center for Applied Mechanics, State Key Laboratory for Strength and Vibration of Mechanical Structures, Xi'an Jiaotong University, Xi'an, China
| | - Xi-Qiao Feng
- AML, Department of Engineering Mechanics, Institute of Biomechanics and Medical Engineering, Tsinghua University, Beijing, China; Center for Nano and Micro Mechanics, Tsinghua University, Beijing, China.
| |
Collapse
|
29
|
Pandya P, Orgaz JL, Sanz-Moreno V. Actomyosin contractility and collective migration: may the force be with you. Curr Opin Cell Biol 2017; 48:87-96. [PMID: 28715714 PMCID: PMC6137077 DOI: 10.1016/j.ceb.2017.06.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/01/2017] [Accepted: 06/23/2017] [Indexed: 01/21/2023]
Abstract
Collective migration relies on the ability of a multicellular co-ordinated unit to efficiently respond to physical changes in their surrounding matrix. Conversely, migrating cohorts physically alter their microenvironment using mechanical forces. During collective migration, actomyosin contractility acts as a central hub coordinating mechanosensing and mechanotransduction responses.
Collective cell migration is essential during physiological processes such as development or wound healing and in pathological conditions such as cancer dissemination. Cells migrating within multicellular tissues experiment different forces which play an intricate role during tissue formation, development and maintenance. How cells are able to respond to these forces depends largely on how they interact with the extracellular matrix. In this review, we focus on mechanics and mechanotransduction in collective migration. In particular, we discuss current knowledge on how cells integrate mechanical signals during collective migration and we highlight actomyosin contractility as a central hub coordinating mechanosensing and mechanotransduction responses.
Collapse
Affiliation(s)
- Pahini Pandya
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Jose L Orgaz
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Victoria Sanz-Moreno
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK.
| |
Collapse
|
30
|
Kurniawan NA, Vos BE, Biebricher A, Wuite GJL, Peterman EJG, Koenderink GH. Fibrin Networks Support Recurring Mechanical Loads by Adapting their Structure across Multiple Scales. Biophys J 2017; 111:1026-34. [PMID: 27602730 DOI: 10.1016/j.bpj.2016.06.034] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/01/2016] [Accepted: 06/29/2016] [Indexed: 12/11/2022] Open
Abstract
Tissues and cells sustain recurring mechanical loads that span a wide range of loading amplitudes and timescales as a consequence of exposure to blood flow, muscle activity, and external impact. Both tissues and cells derive their mechanical strength from fibrous protein scaffolds, which typically have a complex hierarchical structure. In this study, we focus on a prototypical hierarchical biomaterial, fibrin, which is one of the most resilient naturally occurring biopolymers and forms the structural scaffold of blood clots. We show how fibrous networks composed of fibrin utilize irreversible changes in their hierarchical structure at different scales to maintain reversible stress stiffening up to large strains. To trace the origin of this paradoxical resilience, we systematically tuned the microstructural parameters of fibrin and used a combination of optical tweezers and fluorescence microscopy to measure the interactions of single fibrin fibers for the first time, to our knowledge. We demonstrate that fibrin networks adapt to moderate strains by remodeling at the network scale through the spontaneous formation of new bonds between fibers, whereas they adapt to high strains by plastic remodeling of the fibers themselves. This multiscale adaptation mechanism endows fibrin gels with the remarkable ability to sustain recurring loads due to shear flows and wound stretching. Our findings therefore reveal a microscopic mechanism by which tissues and cells can balance elastic nonlinearity and plasticity, and thus can provide microstructural insights into cell-driven remodeling of tissues.
Collapse
Affiliation(s)
- Nicholas A Kurniawan
- Department of Systems Biophysics, FOM Institute AMOLF, Amsterdam, The Netherlands; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands; Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Bart E Vos
- Department of Systems Biophysics, FOM Institute AMOLF, Amsterdam, The Netherlands
| | - Andreas Biebricher
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gijs J L Wuite
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erwin J G Peterman
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Gijsje H Koenderink
- Department of Systems Biophysics, FOM Institute AMOLF, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
van Haaften EE, Bouten CVC, Kurniawan NA. Vascular Mechanobiology: Towards Control of In Situ Regeneration. Cells 2017; 6:E19. [PMID: 28671618 PMCID: PMC5617965 DOI: 10.3390/cells6030019] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 06/16/2017] [Accepted: 06/23/2017] [Indexed: 01/08/2023] Open
Abstract
The paradigm of regenerative medicine has recently shifted from in vitro to in situ tissue engineering: implanting a cell-free, biodegradable, off-the-shelf available scaffold and inducing the development of functional tissue by utilizing the regenerative potential of the body itself. This approach offers a prospect of not only alleviating the clinical demand for autologous vessels but also circumventing the current challenges with synthetic grafts. In order to move towards a hypothesis-driven engineering approach, we review three crucial aspects that need to be taken into account when regenerating vessels: (1) the structure-function relation for attaining mechanical homeostasis of vascular tissues, (2) the environmental cues governing cell function, and (3) the available experimental platforms to test instructive scaffolds for in situ tissue engineering. The understanding of cellular responses to environmental cues leads to the development of computational models to predict tissue formation and maturation, which are validated using experimental platforms recapitulating the (patho)physiological micro-environment. With the current advances, a progressive shift is anticipated towards a rational and effective approach of building instructive scaffolds for in situ vascular tissue regeneration.
Collapse
Affiliation(s)
- Eline E van Haaften
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands.
| |
Collapse
|
32
|
Kurniawan N, van Kempen THS, Sonneveld S, Rosalina TT, Vos BE, Jansen KA, Peters GWM, van de Vosse FN, Koenderink GH. Buffers Strongly Modulate Fibrin Self-Assembly into Fibrous Networks. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:6342-6352. [PMID: 28558246 PMCID: PMC5489959 DOI: 10.1021/acs.langmuir.7b00527] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/27/2017] [Indexed: 05/20/2023]
Abstract
Fibrin is a plasma protein with a central role in blood clotting and wound repair. Upon vascular injury, fibrin forms resilient fibrillar networks (clots) via a multistep self-assembly process, from monomers, to double-stranded protofibrils, to a branched network of thick fibers. In vitro, fibrin self-assembly is sensitive to physicochemical conditions like the solution pH and ionic strength, which tune the strength of the noncovalent driving forces. Here we report a surprising finding that the buffer-which is necessary to control the pH and is typically considered to be inert-also significantly influences fibrin self-assembly. We show by confocal microscopy and quantitative light scattering that various common buffering agents have no effect on the initial assembly of fibrin monomers into protofibrils but strongly hamper the subsequent lateral association of protofibrils into thicker fibers. We further find that the structural changes are independent of the molecular structure of the buffering agents as well as of the activation mechanism and even occur in fibrin networks formed from platelet-poor plasma. This buffer-mediated decrease in protofibril bundling results in a marked reduction in the permeability of fibrin networks but only weakly influences the elastic modulus of fibrin networks, providing a useful tuning parameter to independently control the elastic properties and the permeability of fibrin networks. Our work raises the possibility that fibrin assembly in vivo may be regulated by variations in the acute-phase levels of bicarbonate and phosphate, which act as physiological buffering agents of blood pH. Moreover, our findings add a new example of buffer-induced effects on biomolecular self-assembly to recent findings for a range of proteins and lipids.
Collapse
Affiliation(s)
- Nicholas
A. Kurniawan
- Department
of Systems Biophysics, AMOLF, Amsterdam 1009 DB, The Netherlands
- Department of Biomedical
Engineering & Institute for Complex
Molecular Systems, and Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
| | - Thomas H. S. van Kempen
- Department of Biomedical
Engineering & Institute for Complex
Molecular Systems, and Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
| | - Stijn Sonneveld
- Department
of Systems Biophysics, AMOLF, Amsterdam 1009 DB, The Netherlands
| | - Tilaï T. Rosalina
- Department of Biomedical
Engineering & Institute for Complex
Molecular Systems, and Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
| | - Bart E. Vos
- Department
of Systems Biophysics, AMOLF, Amsterdam 1009 DB, The Netherlands
| | - Karin A. Jansen
- Department
of Systems Biophysics, AMOLF, Amsterdam 1009 DB, The Netherlands
| | - Gerrit W. M. Peters
- Department of Biomedical
Engineering & Institute for Complex
Molecular Systems, and Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
| | - Frans N. van de Vosse
- Department of Biomedical
Engineering & Institute for Complex
Molecular Systems, and Department of Mechanical Engineering, Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
| | - Gijsje H. Koenderink
- Department
of Systems Biophysics, AMOLF, Amsterdam 1009 DB, The Netherlands
- E-mail:
| |
Collapse
|
33
|
Audet-Delage Y, Rouleau M, Rouleau M, Roberge J, Miard S, Picard F, Têtu B, Guillemette C. Cross-Talk between Alternatively Spliced UGT1A Isoforms and Colon Cancer Cell Metabolism. Mol Pharmacol 2017; 91:167-177. [PMID: 28049773 DOI: 10.1124/mol.116.106161] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 12/29/2016] [Indexed: 12/16/2023] Open
Abstract
Alternative splicing at the human glucuronosyltransferase 1 gene locus (UGT1) produces alternate isoforms UGT1A_i2s that control glucuronidation activity through protein-protein interactions. Here, we hypothesized that UGT1A_i2s function as a complex protein network connecting other metabolic pathways with an influence on cancer cell metabolism. This is based on a pathway enrichment analysis of proteomic data that identified several high-confidence candidate interaction proteins of UGT1A_i2 proteins in human tissues-namely, the rate-limiting enzyme of glycolysis pyruvate kinase (PKM), which plays a critical role in cancer cell metabolism and tumor growth. The partnership of UGT1A_i2 and PKM2 was confirmed by coimmunoprecipitation in the HT115 colon cancer cells and was supported by a partial colocalization of these two proteins. In support of a functional role for this partnership, depletion of UGT1A_i2 proteins in HT115 cells enforced the Warburg effect, with a higher glycolytic rate at the expense of mitochondrial respiration, and led to lactate accumulation. Untargeted metabolomics further revealed a significantly altered cellular content of 58 metabolites, including many intermediates derived from the glycolysis and tricarboxylic acid cycle pathways. These metabolic changes were associated with a greater migration potential. The potential relevance of our observations is supported by the down-regulation of UGT1A_i2 mRNA in colon tumors compared with normal tissues. Alternate UGT1A variants may thus be part of the expanding compendium of metabolic pathways involved in cancer biology directly contributing to the oncogenic phenotype of colon cancer cells. Findings uncover new aspects of UGT functions diverging from their transferase activity.
Collapse
Affiliation(s)
- Yannick Audet-Delage
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Michèle Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Mélanie Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Joannie Roberge
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Stéphanie Miard
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Frédéric Picard
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Bernard Têtu
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy (Y.A.-D., Mi.R., Me.R., J.R., C.G.), Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (S.M., F.P.), and Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine (B.T.), Laval University, Québec, Canada
| |
Collapse
|
34
|
Pandya P, Orgaz JL, Sanz-Moreno V. Modes of invasion during tumour dissemination. Mol Oncol 2016; 11:5-27. [PMID: 28085224 PMCID: PMC5423224 DOI: 10.1002/1878-0261.12019] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/24/2016] [Accepted: 10/28/2016] [Indexed: 02/06/2023] Open
Abstract
Cancer cell migration and invasion underlie metastatic dissemination, one of the major problems in cancer. Tumour cells exhibit a striking variety of invasion strategies. Importantly, cancer cells can switch between invasion modes in order to cope with challenging environments. This ability to switch migratory modes or plasticity highlights the challenges behind antimetastasis therapy design. In this Review, we present current knowledge on different tumour invasion strategies, the determinants controlling plasticity and arising therapeutic opportunities. We propose that targeting master regulators controlling plasticity is needed to hinder tumour dissemination and metastasis.
Collapse
Affiliation(s)
- Pahini Pandya
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Jose L Orgaz
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Victoria Sanz-Moreno
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, UK
| |
Collapse
|
35
|
Portillo-Lara R, Annabi N. Microengineered cancer-on-a-chip platforms to study the metastatic microenvironment. LAB ON A CHIP 2016; 16:4063-4081. [PMID: 27605305 DOI: 10.1039/c6lc00718j] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
More than 90% of cancer-related deaths can be attributed to the occurrence of metastatic diseases. Recent studies have highlighted the importance of the multicellular, biochemical and biophysical stimuli from the tumor microenvironment during carcinogenesis, treatment failure, and metastasis. Therefore, there is a need for experimental platforms that are able to recapitulate the complex pathophysiological features of the metastatic microenvironment. Recent advancements in biomaterials, microfluidics, and tissue engineering have led to the development of living multicellular microculture systems, which are maintained in controllable microenvironments and function with organ level complexity. The applications of these "on-chip" technologies for detection, separation, characterization and three dimensional (3D) propagation of cancer cells have been extensively reviewed in previous works. In this contribution, we focus on integrative microengineered platforms that allow the study of multiple aspects of the metastatic microenvironment, including the physicochemical cues from the tumor associated stroma, the heterocellular interactions that drive trans-endothelial migration and angiogenesis, the environmental stresses that metastatic cancer cells encounter during migration, and the physicochemical gradients that direct cell motility and invasion. We discuss the application of these systems as in vitro assays to elucidate fundamental mechanisms of cancer metastasis, as well as their use as human relevant platforms for drug screening in biomimetic microenvironments. We then conclude with our commentaries on current progress and future perspectives of microengineered systems for fundamental and translational cancer research.
Collapse
Affiliation(s)
- R Portillo-Lara
- Department of Chemical Engineering, Northeastern University, 451 Snell Engineering Building, 360 Huntington Ave, Boston, MA 02115, USA. and Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Mexico
| | - N Annabi
- Department of Chemical Engineering, Northeastern University, 451 Snell Engineering Building, 360 Huntington Ave, Boston, MA 02115, USA. and Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|