1
|
Doherty EL, Krohn G, Warren EC, Patton A, Whitworth CP, Rathod M, Biehl A, Aw WY, Freytes DO, Polacheck WJ. Human Cell-Derived Matrix Composite Hydrogels with Diverse Composition for Use in Vasculature-on-chip Models. Adv Healthc Mater 2024; 13:e2400192. [PMID: 38518808 PMCID: PMC11281875 DOI: 10.1002/adhm.202400192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Microphysiological and organ-on-chip platforms seek to address critical gaps in human disease models and drug development that underlie poor rates of clinical success for novel interventions. While the fabrication technology and model cells used to synthesize organs-on-chip have advanced considerably, most platforms rely on animal-derived or synthetic extracellular matrix as a cell substrate, limiting mimicry of human physiology and precluding use in modeling diseases in which matrix dynamics play a role in pathogenesis. Here, the development of human cell-derived matrix (hCDM) composite hydrogels for use in 3D microphysiologic models of the vasculature is reported. hCDM composite hydrogels are derived from human donor fibroblasts and maintain a complex milieu of basement membrane, proteoglycans, and nonfibrillar matrix components. The use of hCDM composite hydrogels as 2D and 3D cell culture substrates is demonstrated, and hCDM composite hydrogels are patterned to form engineered human microvessels. Interestingly, hCDM composite hydrogels are enriched in proteins associated with vascular morphogenesis as determined by mass spectrometry, and functional analysis demonstrates proangiogenic signatures in human endothelial cells cultured in these hydrogels. In conclusion, this study suggests that human donor-derived hCDM composite hydrogels could address technical gaps in human organs-on-chip development and serve as substrates to promote vascularization.
Collapse
Affiliation(s)
- Elizabeth L Doherty
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Grace Krohn
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Emily C Warren
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Alexandra Patton
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Chloe P Whitworth
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill School of Medicine, 130 Mason Farm Road, Chapel Hill, Carolina, NC 27599, USA
| | - Mitesh Rathod
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Andreea Biehl
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Wen Yih Aw
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - Donald O Freytes
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
| | - William J Polacheck
- The Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, 10010 Mary Ellen Jones Building, 116 Manning Drive, Chapel Hill, NC 27514, USA
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, 111 Mason Farm Road, Chapel Hill, Carolina, NC 27599, USA
| |
Collapse
|
2
|
Fathi P, Sundaresan V, Alfonso AL, Rama Varma A, Sadtler K. Factors Affecting the Evaluation of Collagen Deposition and Fibrosis In Vitro. Tissue Eng Part A 2024; 30:367-380. [PMID: 38511512 PMCID: PMC11250831 DOI: 10.1089/ten.tea.2023.0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/30/2024] [Indexed: 03/22/2024] Open
Abstract
Immune responses to biomedical implants, wound healing, and diseased tissues often involve collagen deposition by fibroblasts and other stromal cells. Dysregulated collagen deposition can lead to complications, such as biomaterial fibrosis, cardiac fibrosis, desmoplasia, liver fibrosis, and pulmonary fibrosis, which can ultimately result in losses of organ function or failure of biomedical implants. Current in vitro methods to induce collagen deposition include growing the cells under macromolecular crowding conditions or on fibronectin-coated surfaces. However, the majority of these methods have been demonstrated with a single cell line, and the combined impacts of culture conditions and postculture processing on collagen deposition have not been explored in detail. In this work, the effects of macromolecular crowding versus fibronectin coating, fixation with methanol versus fixation with paraformaldehyde, and use of plastic substrates versus glass substrates were evaluated using the WI-38 human lung fibroblast cell line. Fibronectin coating was found to provide enhanced collagen deposition under macromolecular crowding conditions, while a higher plating density led to improved collagen I deposition compared with macromolecular crowding. Collagen deposition was found to be more apparent on plastic substrates than on glass substrates. The effects of primary cells versus cell lines, and mouse cells versus human cells, were evaluated using WI-38 cells, primary human lung fibroblasts, primary human dermal fibroblasts, primary mouse lung fibroblasts, primary mouse dermal fibroblasts, and the L929 mouse fibroblast cell line. Cell lines exhibited enhanced collagen I deposition compared with primary cells. Furthermore, collagen deposition was quantified with picrosirius red staining, and plate-based drug screening through picrosirius red staining of decellularized extracellular matrices was demonstrated. The results of this study provide detailed conditions under which collagen deposition can be induced in vitro in multiple cell types, with applications including material development, development of potential antifibrotic therapies, and mechanistic investigation of disease pathways. Impact Statement This study demonstrated the effects of cell type, biological conditions, fixative, culture substrate, and staining method on in vitro collagen deposition and visualization. Further the utility of plate-based picrosirius red staining of decellularized extracellular matrices for drug screening through collagen quantification was demonstrated. These results should provide clarity and a path forward for researchers who aim to conduct in vitro experiments on collagen deposition.
Collapse
Affiliation(s)
- Parinaz Fathi
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
- Unit for NanoEngineering and MicroPhysiological Systems (UNEMPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Vanathi Sundaresan
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Andrea Lucia Alfonso
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Anagha Rama Varma
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
- Unit for NanoEngineering and MicroPhysiological Systems (UNEMPS), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Kaitlyn Sadtler
- Section on Immunoengineering, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
3
|
Marino-Bravante GE, Carey AE, Hüser L, Dixit A, Wang V, Kaur A, Liu Y, Ding S, Schnellmann R, Gerecht S, Gu L, Eisinger-Mathason TSK, Chhabra Y, Weeraratna AT. Age-dependent loss of HAPLN1 erodes vascular integrity via indirect upregulation of endothelial ICAM1 in melanoma. NATURE AGING 2024; 4:350-363. [PMID: 38472454 DOI: 10.1038/s43587-024-00581-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 01/26/2024] [Indexed: 03/14/2024]
Abstract
Melanoma, the most lethal form of skin cancer, often has worse outcomes in older patients. We previously demonstrated that an age-related decrease in the secreted extracellular matrix (ECM) protein HAPLN1 has a role in slowing melanoma progression. Here we show that HAPLN1 in the dermal ECM is sufficient to maintain the integrity of melanoma-associated blood vessels, as indicated by increased collagen and VE-cadherin expression. Specifically, we show that HAPLN1 in the ECM increases hyaluronic acid and decreases endothelial cell expression of ICAM1. ICAM1 phosphorylates and internalizes VE-cadherin, a critical determinant of vascular integrity, resulting in permeable blood vessels. We found that blocking ICAM1 reduces tumor size and metastasis in older mice. These results suggest that HAPLN1 alters endothelial ICAM1expression in an indirect, matrix-dependent manner. Targeting ICAM1 could be a potential treatment strategy for older patients with melanoma, emphasizing the role of aging in tumorigenesis.
Collapse
Affiliation(s)
- Gloria E Marino-Bravante
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Alexis E Carey
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Laura Hüser
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Agrani Dixit
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Vania Wang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Amanpreet Kaur
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Liu
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Supeng Ding
- Department of Materials Science and Engineering, Johns Hopkins Whiting School of Engineering, Baltimore, MD, USA
| | - Rahel Schnellmann
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins Whiting School of Engineering, Baltimore, MD, USA
| | - T S Karin Eisinger-Mathason
- Abramson Family Cancer Research Institute, Department of Pathology & Laboratory Medicine, Penn Sarcoma Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yash Chhabra
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Cancer Signaling and Microenvironment, FoxChase Cancer Center, Philadelphia, PA, USA.
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
4
|
Jin Q, Pandey D, Thompson CB, Lewis S, Sung HW, Nguyen TD, Kuo S, Wilson KL, Gracias DH, Romer LH. Acute downregulation of emerin alters actomyosin cytoskeleton connectivity and function. Biophys J 2023; 122:3690-3703. [PMID: 37254483 PMCID: PMC10541481 DOI: 10.1016/j.bpj.2023.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/30/2023] [Accepted: 05/22/2023] [Indexed: 06/01/2023] Open
Abstract
Fetal lung fibroblasts contribute dynamic infrastructure for the developing lung. These cells undergo dynamic mechanical transitions, including cyclic stretch and spreading, which are integral to lung growth in utero. We investigated the role of the nuclear envelope protein emerin in cellular responses to these dynamic mechanical transitions. In contrast to control cells, which briskly realigned their nuclei, actin cytoskeleton, and extracellular matrices in response to cyclic stretch, fibroblasts that were acutely downregulated for emerin showed incomplete reorientation of both nuclei and actin cytoskeleton. Emerin-downregulated fibroblasts were also aberrantly circular in contrast to the spindle-shaped controls and exhibited an altered pattern of filamentous actin organization that was disconnected from the nucleus. Emerin knockdown was also associated with reduced myosin light chain phosphorylation during cell spreading. Interestingly, emerin-downregulated fibroblasts also demonstrated reduced fibronectin fibrillogenesis and production. These findings indicate that nuclear-cytoskeletal coupling serves a role in the dynamic regulation of cytoskeletal structure and function and may also impact the transmission of traction force to the extracellular matrix microenvironment.
Collapse
Affiliation(s)
- Qianru Jin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Deepesh Pandey
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Carol B Thompson
- Biostatistics Center, Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Shawna Lewis
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Hyun Woo Sung
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Thao D Nguyen
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Scot Kuo
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland; Microscope Facility, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Katherine L Wilson
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - David H Gracias
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland; Center for MicroPhysiological Systems, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland; Department of Chemistry, Johns Hopkins University, Baltimore, Maryland; Laboratory for Computational Sensing and Robotics, Johns Hopkins University, Baltimore, Maryland
| | - Lewis H Romer
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, Maryland; Center for Cell Dynamics, Johns Hopkins School of Medicine, Baltimore, Maryland.
| |
Collapse
|
5
|
Collagen-Based Biomimetic Systems to Study the Biophysical Tumour Microenvironment. Cancers (Basel) 2022; 14:cancers14235939. [PMID: 36497421 PMCID: PMC9739814 DOI: 10.3390/cancers14235939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/22/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022] Open
Abstract
The extracellular matrix (ECM) is a pericellular network of proteins and other molecules that provides mechanical support to organs and tissues. ECM biophysical properties such as topography, elasticity and porosity strongly influence cell proliferation, differentiation and migration. The cell's perception of the biophysical microenvironment (mechanosensing) leads to altered gene expression or contractility status (mechanotransduction). Mechanosensing and mechanotransduction have profound implications in both tissue homeostasis and cancer. Many solid tumours are surrounded by a dense and aberrant ECM that disturbs normal cell functions and makes certain areas of the tumour inaccessible to therapeutic drugs. Understanding the cell-ECM interplay may therefore lead to novel and more effective therapies. Controllable and reproducible cell culturing systems mimicking the ECM enable detailed investigation of mechanosensing and mechanotransduction pathways. Here, we discuss ECM biomimetic systems. Mainly focusing on collagen, we compare and contrast structural and molecular complexity as well as biophysical properties of simple 2D substrates, 3D fibrillar collagen gels, cell-derived matrices and complex decellularized organs. Finally, we emphasize how the integration of advanced methodologies and computational methods with collagen-based biomimetics will improve the design of novel therapies aimed at targeting the biophysical and mechanical features of the tumour ECM to increase therapy efficacy.
Collapse
|
6
|
Guo T, He C, Venado A, Zhou Y. Extracellular Matrix Stiffness in Lung Health and Disease. Compr Physiol 2022; 12:3523-3558. [PMID: 35766837 PMCID: PMC10088466 DOI: 10.1002/cphy.c210032] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The extracellular matrix (ECM) provides structural support and imparts a wide variety of environmental cues to cells. In the past decade, a growing body of work revealed that the mechanical properties of the ECM, commonly known as matrix stiffness, regulate the fundamental cellular processes of the lung. There is growing appreciation that mechanical interplays between cells and associated ECM are essential to maintain lung homeostasis. Dysregulation of ECM-derived mechanical signaling via altered mechanosensing and mechanotransduction pathways is associated with many common lung diseases. Matrix stiffening is a hallmark of lung fibrosis. The stiffened ECM is not merely a sequelae of lung fibrosis but can actively drive the progression of fibrotic lung disease. In this article, we provide a comprehensive view on the role of matrix stiffness in lung health and disease. We begin by summarizing the effects of matrix stiffness on the function and behavior of various lung cell types and on regulation of biomolecule activity and key physiological processes, including host immune response and cellular metabolism. We discuss the potential mechanisms by which cells probe matrix stiffness and convert mechanical signals to regulate gene expression. We highlight the factors that govern matrix stiffness and outline the role of matrix stiffness in lung development and the pathogenesis of pulmonary fibrosis, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. We envision targeting of deleterious matrix mechanical cues for treatment of fibrotic lung disease. Advances in technologies for matrix stiffness measurements and design of stiffness-tunable matrix substrates are also explored. © 2022 American Physiological Society. Compr Physiol 12:3523-3558, 2022.
Collapse
Affiliation(s)
- Ting Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA.,Department of Respiratory Medicine, the Second Xiangya Hospital, Central-South University, Changsha, Hunan, China
| | - Chao He
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Aida Venado
- Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
7
|
Pahapale GJ, Tao J, Nikolic M, Gao S, Scarcelli G, Sun SX, Romer LH, Gracias DH. Directing Multicellular Organization by Varying the Aspect Ratio of Soft Hydrogel Microwells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104649. [PMID: 35434926 PMCID: PMC9189654 DOI: 10.1002/advs.202104649] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/08/2022] [Indexed: 06/03/2023]
Abstract
Multicellular organization with precise spatial definition is essential to various biological processes, including morphogenesis, development, and healing in vascular and other tissues. Gradients and patterns of chemoattractants are well-described guides of multicellular organization, but the influences of 3D geometry of soft hydrogels are less well defined. Here, the discovery of a new mode of endothelial cell self-organization guided by combinatorial effects of stiffness and geometry, independent of protein or chemical patterning, is described. Endothelial cells in 2 kPa microwells are found to be ≈30 times more likely to migrate to the edge to organize in ring-like patterns than in stiff 35 kPa microwells. This organization is independent of curvature and significantly more pronounced in 2 kPa microwells with aspect ratio (perimeter/depth) < 25. Physical factors of cells and substrates that drive this behavior are systematically investigated and a mathematical model that explains the organization by balancing the dynamic interaction between tangential cytoskeletal tension, cell-cell, and cell-substrate adhesion is presented. These findings demonstrate the importance of combinatorial effects of geometry and stiffness in complex cellular organization that can be leveraged to facilitate the engineering of bionics and integrated model organoid systems with customized nutrient vascular networks.
Collapse
Affiliation(s)
- Gayatri J. Pahapale
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jiaxiang Tao
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Milos Nikolic
- Maryland Biophysics ProgramInstitute for Physical Science and TechnologyUniversity of MarylandCollege ParkMD20742USA
| | - Sammy Gao
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Giuliano Scarcelli
- Maryland Biophysics ProgramInstitute for Physical Science and Technology and Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Sean X. Sun
- Department of Mechanical EngineeringCell Biologyand Institute of NanoBioTechnology (INBT)Johns Hopkins UniversityBaltimoreMD21218USA
| | - Lewis H. Romer
- Department of Cell BiologyAnesthesiology and Critical Care MedicineBiomedical EngineeringPediatricsand Center for Cell DynamicsJohns Hopkins School of MedicineBaltimoreMD21205USA
| | - David H. Gracias
- Department of Chemical and Biomolecular EngineeringMaterials Science and EngineeringChemistry and Laboratory for Computational Sensing and Robotics (LCSR)Johns Hopkins UniversityBaltimoreMD21218USA
- Department of Oncology and Sidney Kimmel Comprehensive Cancer CenterJohns Hopkins School of MedicineBaltimoreMD21205USA
| |
Collapse
|
8
|
Satyam A, Tsokos MG, Tresback JS, Zeugolis DI, Tsokos GC. Cell derived extracellular matrix-rich biomimetic substrate supports podocyte proliferation, differentiation and maintenance of native phenotype. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1908752. [PMID: 33692659 PMCID: PMC7939063 DOI: 10.1002/adfm.201908752] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Indexed: 06/12/2023]
Abstract
Current technologies and available scaffold materials do not support long-term cell viability, differentiation and maintenance of podocytes, the ultra-specialized kidney resident cells that are responsible for the filtration of the blood. We developed a new platform which imitates the native kidney microenvironment by decellularizing fibroblasts grown on surfaces with macromolecular crowding. Human immortalized podocytes cultured on this platform displayed superior viability and metabolic activity up to 28 days compared to podocytes cultured on tissue culture plastic surfaces. The new platform displayed a softer surface and an abundance of growth factors and associated molecules. More importantly it enabled podocytes to display molecules responsible for their structure and function and a superior development of intercellular connections/interdigitations, consistent with maturation. The new platform can be used to study podocyte biology, test drug toxicity and determine whether sera from patients with podocytopathies are involved in the expression of glomerular pathology.
Collapse
Affiliation(s)
- Abhigyan Satyam
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, United States
| | - Maria G Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, United States
| | - Jason S Tresback
- Center for Nanoscale Systems, Laboratory for Integrated Science and Engineering, Harvard University, Cambridge, MA, 02138, United States
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Centre for Research in Medical Devices (CURAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, United States
| |
Collapse
|
9
|
Lee BJ, Hegewisch Solloa E, Shannon MJ, Mace EM. Generation of cell-derived matrices that support human NK cell migration and differentiation. J Leukoc Biol 2020; 108:1369-1378. [PMID: 32392635 DOI: 10.1002/jlb.1ma0420-635r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/16/2020] [Accepted: 04/26/2020] [Indexed: 12/20/2022] Open
Abstract
Human NK cells are effectors of the innate immune system that originate from hematopoietic precursors in the bone marrow. While stromal cell lines that support NK cell development from hematopoietic precursors are often used to generate mature NK cells from lymphoid precursors in vitro, the nature of contributing factors of these stromal cells to the generation of functionally mature NK cells has been poorly described. Previous studies have shown that developing NK cells adhere to, and migrate on, developmentally supportive stroma. Here, we describe the generation of cell-derived matrices (CDMs) from a commonly used murine fetal liver stromal cell line. These CDMs are derived directly from the same EL08.1D2 stromal cell line known to support NK cell differentiation and contain ECM structural components fibronectin and collagen. We demonstrate that CDMs support NK cell adhesion and migration with similar properties as intact cells. Further, we show that CDMs support NK cell maturation from lymphoid precursors in vitro, albeit with reduced cell survival compared to intact cell-based differentiation. Together, these results describe a cell-free system that supports NK cell development and that can serve as a useful model for studying the nature of the biochemical interactions between NK cell developmental intermediates and developmentally supportive substrates.
Collapse
Affiliation(s)
- Barclay J Lee
- Department of Bioengineering, Rice University, Houston, Texas, USA.,Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Everardo Hegewisch Solloa
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Michael J Shannon
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Emily M Mace
- Department of Pediatrics, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
10
|
Cooper JG, Sicard D, Sharma S, Van Gulden S, McGuire TL, Cajiao MP, Tschumperlin DJ, Kessler JA. Spinal Cord Injury Results in Chronic Mechanical Stiffening. J Neurotrauma 2020; 37:494-506. [PMID: 31516087 PMCID: PMC6978780 DOI: 10.1089/neu.2019.6540] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Gliosis and fibrosis after spinal cord injury (SCI) lead to formation of a scar that is thought to present both molecular and mechanical barriers to neuronal regeneration. The scar consists of a meshwork of reactive glia and deposited, cross-linked, extracellular matrix (ECM) that has long been assumed to present a mechanically "stiff" blockade. However, remarkably little quantitative information is available about the rheological properties of chronically injured spinal tissue. In this study we utilize atomic force microscopy microindentation to provide quantitative evidence of chronic mechanical stiffening after SCI. Using the results of this tissue characterization, we assessed the sensitivity of both mouse and human astrocytes in vitro and determined that they are exquisitely mechanosensitive within the relevant range of substrate stiffness observed in the injured/uninjured spinal cord. We then utilized a novel immune modifying nanoparticle (IMP) treatment as a tool to reveal fibrotic scarring as one of the key drivers of mechanical stiffening after SCI in vivo. We also demonstrate that glial scar-forming astrocytes form a highly aligned, anisotropic network of glial fibers after SCI, and that IMP treatment mitigates this pathological alignment. Taken together, our results identify chronic mechanical stiffening as a critically important aspect of the complex lesion milieu after SCI that must be considered when assessing and developing potential clinical interventions for SCI.
Collapse
Affiliation(s)
- John G. Cooper
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, College of Medicine and Science, Mayo Clinic, Rochester, Minnesota
| | - Sripadh Sharma
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Stephanie Van Gulden
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Tammy L. McGuire
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Miguel Pareja Cajiao
- Department of Anesthesiology, College of Medicine and Science, Mayo Clinic, Rochester, Minnesota
| | - Daniel J. Tschumperlin
- Department of Physiology and Biomedical Engineering, College of Medicine and Science, Mayo Clinic, Rochester, Minnesota
| | - John A. Kessler
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
11
|
Godeau AL, Delanoë-Ayari H, Riveline D. Generation of fluorescent cell-derived-matrix to study 3D cell migration. Methods Cell Biol 2020; 156:185-203. [PMID: 32222219 DOI: 10.1016/bs.mcb.2019.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Cell migration is involved in key phenomena in biology, ranging from development to cancer. Fibroblasts move between organs in 3D polymeric networks. So far, motile cells were mainly tracked in vitro on Petri dishes or on coverslips, i.e., 2D flat surfaces, which made the extrapolation to 3D physiological environments difficult. We therefore prepared 3D Cell Derived Matrices (CDM) with specific characteristics with the goal of extracting the main readouts required to measure and characterize cell motion: cell specific matrix deformation through the tracking of fluorescent fibronectin within CDM, focal contacts as the cell anchor and acto-myosin cytoskeleton which applies cellular forces. We report our method for generating this assay of physiological-like gel with relevant readouts together with its potential impact in explaining cell motility in vivo.
Collapse
Affiliation(s)
- Amélie Luise Godeau
- Laboratory of Cell Physics ISIS/IGBMC, CNRS and University of Strasbourg, Strasbourg, France; Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France
| | - Hélène Delanoë-Ayari
- University of Lyon, Université Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, Villeurbanne, France
| | - Daniel Riveline
- Laboratory of Cell Physics ISIS/IGBMC, CNRS and University of Strasbourg, Strasbourg, France; Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France; Centre National de la Recherche Scientifique, UMR7104, Illkirch, France; Institut National de la Santé et de la Recherche Médicale, U964, Illkirch, France; Université de Strasbourg, Illkirch, France.
| |
Collapse
|
12
|
Keurhorst D, Liashkovich I, Frontzek F, Nitzlaff S, Hofschröer V, Dreier R, Stock C. MMP3 activity rather than cortical stiffness determines NHE1-dependent invasiveness of melanoma cells. Cancer Cell Int 2019; 19:285. [PMID: 31728131 PMCID: PMC6842528 DOI: 10.1186/s12935-019-1015-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/01/2019] [Indexed: 12/31/2022] Open
Abstract
Background Both cell adhesion and matrix metalloproteinase (MMP) activity depend on pH at the cell surface. By regulating extracellular juxtamembrane pH, the Na+/H+ exchanger NHE1 plays a significant part in human melanoma (MV3) cell migration and invasion. Because NHE1, besides its pH-regulatory transport function, also serves as a structural element tying the cortical actin cytoskeleton to the plasma membrane, we investigated whether NHE1 affects cortical stiffness of MV3 cells, and how this makes an impact on their invasiveness. Methods NHE1 overexpressing MV3 cells were compared to the corresponding mock-transfected control cells. NHE1 expression was verified by Western blotting, cariporide (HOE642) was used to inhibit NHE1 activity, cell stiffness was determined by atomic force microscopy, and F-actin was visualized by phalloidin-staining. Migration on, and invasion of, native and glutaraldehyde-fixed collagen I substrates were analyzed using time-lapse video microscopy and Boyden-chamber assays, respectively. MMP secretion and activity were detected by Western blot and zymography, respectively. MMP activity was inhibited with NNGH. Results The cortical, but not the bulk stiffness, was significantly higher in NHE1 overexpressing cells. This increase in cortical stiffness was accompanied by a reorganization of the cortical cytoskeleton, i.e. a condensation of F-actin underneath and along the plasma membrane. However, it was not affected by NHE1 inhibition. Nevertheless, actin dynamics is required for cell invasion as demonstrated with the application of cytochalasin D. NHE1 overexpression was associated with an elevated MMP3 secretion and an increase in the invasion of a native matrix. This increase in invasiveness could be antagonized by the MMP inhibitor NNGH. Transmigration through a glutaraldehyde-fixed, indigestible substrate was not affected by NHE1 overexpression. Conclusion NHE1, as a structural element and independently of its transport activity, contributes to the organization of the cortical F-actin meshwork and thus impacts cortical stiffness. Since NHE1 overexpression stimulates MMP3 secretion but does not change transmigration through a fixed substrate, MV3 cell invasion of a native substrate depends on MMP activity rather than on a modifiable cortical stiffness.
Collapse
Affiliation(s)
- Dennis Keurhorst
- 1Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Ivan Liashkovich
- 1Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Fabian Frontzek
- 2Department of Oncology and Hematology, University Hospital of Münster, Albert-Schweitzer-Campus 1, 48149 Münster, Germany
| | - Svenja Nitzlaff
- 3Institute of Animal Physiology, University of Münster, Schlossplatz 8, 48143 Münster, Germany
| | - Verena Hofschröer
- 1Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany
| | - Rita Dreier
- 4Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyer-Str. 15, 48149 Münster, Germany
| | - Christian Stock
- 1Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149 Münster, Germany.,5Department of Gastroenterology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
13
|
Pahapale GJ, Gao S, Romer LH, Gracias DH. Hierarchically Curved Gelatin for 3D Biomimetic Cell Culture. ACS APPLIED BIO MATERIALS 2019; 2:6004-6011. [DOI: 10.1021/acsabm.9b00916] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
14
|
Hoshiba T. Decellularized Extracellular Matrix for Cancer Research. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E1311. [PMID: 31013621 PMCID: PMC6515435 DOI: 10.3390/ma12081311] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 12/11/2022]
Abstract
Genetic mutation and alterations of intracellular signaling have been focused on to understand the mechanisms of oncogenesis and cancer progression. Currently, it is pointed out to consider cancer as tissues. The extracellular microenvironment, including the extracellular matrix (ECM), is important for the regulation of cancer cell behavior. To comprehensively investigate ECM roles in the regulation of cancer cell behavior, decellularized ECM (dECM) is now used as an in vitro ECM model. In this review, I classify dECM with respect to its sources and summarize the preparation and characterization methods for dECM. Additionally, the examples of cancer research using the dECM were introduced. Finally, future perspectives of cancer studies with dECM are described in the conclusions.
Collapse
Affiliation(s)
- Takashi Hoshiba
- Biotechnology Group, Tokyo Metropolitan Industrial Technology Research Institute, Koto-ku, Tokyo 135-0064, Japan.
- Research Center for Functional Materials, National Institute for Materials Science, Tsukuba 305-0044, Ibaraki, Japan.
| |
Collapse
|
15
|
Du P, Casavitri C, Suhaeri M, Wang PY, Lee JH, Koh WG, Park K. A Fibrous Hybrid Patch Couples Cell-Derived Matrix and Poly(l-lactide-co-caprolactone) for Endothelial Cells Delivery and Skin Wound Repair. ACS Biomater Sci Eng 2018; 5:900-910. [DOI: 10.1021/acsbiomaterials.8b01118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Ping Du
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Center for Human Tissues & Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Cininta Casavitri
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Muhammad Suhaeri
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Peng-Yuan Wang
- Center for Human Tissues & Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jong Ho Lee
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| |
Collapse
|
16
|
Ani CJ, Obayemi JD, Uzonwanne VO, Danyuo Y, Odusanya OS, Hu J, Malatesta K, Soboyejo WO. A shear assay study of single normal/breast cancer cell deformation and detachment from poly-di-methyl-siloxane (PDMS) surfaces. J Mech Behav Biomed Mater 2018; 91:76-90. [PMID: 30544025 DOI: 10.1016/j.jmbbm.2018.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/25/2018] [Accepted: 11/12/2018] [Indexed: 02/01/2023]
Abstract
This paper presents the results of a combined experimental and analytical/computational study of viscoelastic cell deformation and detachment from poly-di-methyl-siloxane (PDMS) surfaces. Fluid mechanics and fracture mechanics concepts are used to model the detachment of biological cells observed under shear assay conditions. The analytical and computational models are used to compute crack driving forces, which are then related to crack extension during the detachment of normal breast cells and breast cancer cells from PDMS surfaces that are relevant to biomedical implants. The interactions between cells and the extracellular matrix, or the extracellular matrix and the PDMS substrate, are then characterized using force microscopy measurements of the pull-off forces that are used to determine the adhesion energies. Finally, fluorescence microscopy staining of the cytosketelal structures (actin, micro-tubulin and cyto-keratin), transmembrane proteins (vimentin) and the ECM structures (Arginin Glycine Aspartate - RGD) is used to show that the detachment of cells during the shear assay experiments occurs via interfacial cracking between (between the ECM and the cell membranes) with a high incidence of crack bridging by transmembrane vinculin structures that undergo pull-out until they detach from the actin cytoskeletal structure. The implications of the results are discussed for the design of interfaces that are relevant to implantable biomedical devices and normal/cancer tissue.
Collapse
Affiliation(s)
- C J Ani
- Department of Theoretical and Applied Physics, African University of Science and Technology, Km 10, Airport Road, Galadimawa, Abuja, Federal Capital Territory, Nigeria; Department of Physics, Salem University, Km 16, PMB 1060, Lokoja, Kogi State, Nigeria
| | - J D Obayemi
- Department of Mechanical Engineering, Worcester Polytechnic Institute (WPI), Worcester, MA 01609, USA
| | - V O Uzonwanne
- Department of Mechanical Engineering, Worcester Polytechnic Institute (WPI), Worcester, MA 01609, USA
| | - Y Danyuo
- Department of Mechanical Engineering, Ashesi University, Berekuso, Ghana; Department of Materials Science and Engineering, African University of Science and Technology, Km 10, Airport Road, Galadimawa, Abuja, Federal Capital Territory, Nigeria
| | - O S Odusanya
- Advanced Biotechnology Laboratory, Sheda Science and Technology Complex, Abuja, Nigeria
| | - J Hu
- Princeton Institute for the Science and Technology of Materials (PRISM), and The Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
| | - K Malatesta
- Princeton Institute for the Science and Technology of Materials (PRISM), and The Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, NJ 08544, USA
| | - W O Soboyejo
- Department of Mechanical Engineering, Worcester Polytechnic Institute (WPI), Worcester, MA 01609, USA; Department of Materials Science and Engineering, African University of Science and Technology, Km 10, Airport Road, Galadimawa, Abuja, Federal Capital Territory, Nigeria; Advanced Biotechnology Laboratory, Sheda Science and Technology Complex, Abuja, Nigeria.
| |
Collapse
|
17
|
Michalik M, Wójcik-Pszczoła K, Paw M, Wnuk D, Koczurkiewicz P, Sanak M, Pękala E, Madeja Z. Fibroblast-to-myofibroblast transition in bronchial asthma. Cell Mol Life Sci 2018; 75:3943-3961. [PMID: 30101406 PMCID: PMC6182337 DOI: 10.1007/s00018-018-2899-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 07/26/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
Abstract
Bronchial asthma is a chronic inflammatory disease in which bronchial wall remodelling plays a significant role. This phenomenon is related to enhanced proliferation of airway smooth muscle cells, elevated extracellular matrix protein secretion and an increased number of myofibroblasts. Phenotypic fibroblast-to-myofibroblast transition represents one of the primary mechanisms by which myofibroblasts arise in fibrotic lung tissue. Fibroblast-to-myofibroblast transition requires a combination of several types of factors, the most important of which are divided into humoural and mechanical factors, as well as certain extracellular matrix proteins. Despite intensive research on the nature of this process, its underlying mechanisms during bronchial airway wall remodelling in asthma are not yet fully clarified. This review focuses on what is known about the nature of fibroblast-to-myofibroblast transition in asthma. We aim to consider possible mechanisms and conditions that may play an important role in fibroblast-to-myofibroblast transition but have not yet been discussed in this context. Recent studies have shown that some inherent and previously undescribed features of fibroblasts can also play a significant role in fibroblast-to-myofibroblast transition. Differences observed between asthmatic and non-asthmatic bronchial fibroblasts (e.g., response to transforming growth factor β, cell shape, elasticity, and protein expression profile) may have a crucial influence on this phenomenon. An accurate understanding and recognition of all factors affecting fibroblast-to-myofibroblast transition might provide an opportunity to discover efficient methods of counteracting this phenomenon.
Collapse
Affiliation(s)
- Marta Michalik
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Wójcik-Pszczoła
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| | - Milena Paw
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Dawid Wnuk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Paulina Koczurkiewicz
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Marek Sanak
- Division of Molecular Biology and Clinical Genetics, Department of Medicine, Jagiellonian University Medical College, Skawińska 8, 31-066, Kraków, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| |
Collapse
|
18
|
Zhou B, Zhang X. Lung mass density analysis using deep neural network and lung ultrasound surface wave elastography. ULTRASONICS 2018; 89:173-177. [PMID: 29852466 PMCID: PMC6014933 DOI: 10.1016/j.ultras.2018.05.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 05/07/2018] [Accepted: 05/22/2018] [Indexed: 06/01/2023]
Abstract
Lung mass density is directly associated with lung pathology. Computed Tomography (CT) evaluates lung pathology using the Hounsfield unit (HU) but not lung density directly. We have developed a lung ultrasound surface wave elastography (LUSWE) technique to measure the surface wave speed of superficial lung tissue. The objective of this study was to develop a method for analyzing lung mass density of superficial lung tissue using a deep neural network (DNN) and synthetic data of wave speed measurements with LUSWE. The synthetic training dataset of surface wave speed, excitation frequency, lung mass density, and viscoelasticity from LUSWE (788,000 in total) was used to train the DNN model. The DNN was composed of 3 hidden layers of 1024 neurons for each layer and trained for 10 epochs with a batch size of 4096 and a learning rate of 0.001 with three types of optimizers. The test dataset (4000) of wave speeds at three excitation frequencies (100, 150, and 200 Hz) and shear elasticity of superficial lung tissue was used to predict the lung density and evaluate its accuracy compared with predefined lung mass densities. This technique was then validated on a sponge phantom experiment. The obtained results showed that predictions matched well with test dataset (validation accuracy is 0.992) and experimental data in the sponge phantom experiment. This method may be useful to analyze lung mass density by using the DNN model together with the surface wave speed and lung stiffness measurements.
Collapse
Affiliation(s)
- Boran Zhou
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA
| | - Xiaoming Zhang
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905, USA.
| |
Collapse
|
19
|
Abstract
The ability to create cell-derived decellularized matrices in a dish gives researchers the opportunity to possess a bioactive, biocompatible material made up of fibrillar proteins and other factors that recapitulates key features of the native structure and composition of in vivo microenvironments. By using cells in a culture system to provide a natural ECM, decellularization allows for a high degree of customization through the introduction of selected proteins and soluble factors. The culture system, culture medium, cell types, and physical environments can be varied to provide specialized ECMs for wide-ranging applications to study cell-ECM signaling, cell migration, cell differentiation, and tissue engineering purposes. This chapter describes a procedure for performing a detergent and high pH-based extraction that leaves the native, cell-assembled ECM intact while removing cellular materials. We address common evaluation methods for assessing the ECM and its composition as well as potential uses for a decellularized ECM.
Collapse
|
20
|
Kaukonen R, Jacquemet G, Hamidi H, Ivaska J. Cell-derived matrices for studying cell proliferation and directional migration in a complex 3D microenvironment. Nat Protoc 2017; 12:2376-2390. [PMID: 29048422 DOI: 10.1038/nprot.2017.107] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
2D surfaces offer simple analysis of cells in culture, yet these often yield different cell morphologies and responses from those observed in vivo. Considerable effort has therefore been expended on the generation of more tissue-like environments for the study of cell behavior in vitro. Purified matrix proteins provide a 3D scaffold that better mimics the in vivo situation; however, these are far removed from the complex tissue composition seen in vivo. Cell-derived matrices (CDMs) offer a more physiologically relevant alternative for studying in vivo-like cell behavior in vitro. In the protocol described here, fibroblasts cultured on gelatin-coated surfaces are maintained in the presence of ascorbic acid to strengthen matrix deposition over 1-3 weeks. The resulting fibrillar CDMs are denuded of cells, and other cells are subsequently cultured on them, after which their behavior is monitored. We also demonstrate how to use CDMs as an in vivo-relevant reductionist model for studying tumor-stroma-induced changes in carcinoma cell proliferation and migration.
Collapse
Affiliation(s)
- Riina Kaukonen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Guillaume Jacquemet
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Biochemistry, University of Turku, Turku, Finland
| |
Collapse
|
21
|
Measured pulmonary arterial tissue stiffness is highly sensitive to AFM indenter dimensions. J Mech Behav Biomed Mater 2017; 74:118-127. [PMID: 28595103 DOI: 10.1016/j.jmbbm.2017.05.039] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 05/22/2017] [Accepted: 05/30/2017] [Indexed: 12/28/2022]
Abstract
The mechanical properties of pulmonary tissues are important in normal function and the development of diseases such as pulmonary arterial hypertension. Hence it is critical to measure lung tissue micromechanical properties as accurately as possible in order to gain insight into the normal and pathological range of tissue stiffness associated with development, aging and disease processes. In this study, we used atomic force microscopy (AFM) micro-indentation to characterize the Young's modulus of small human pulmonary arteries (vessel diameter less than 100µm), and examined the influence of AFM tip geometry and diameter, lung tissue section thickness and the range of working force applied to the sample on the measured modulus. We observed a significant increase of the measured Young's modulus of pulmonary vessels (one order of magnitude) associated with the use of a pyramidal sharp AFM tips (20nm radius), compared to two larger spherical tips (1 and 2.5µm radius) which generated statistically indistinguishable results. The effect of tissue section thickness (ranging from 10 to 50 μm) on the measured elastic modulus was relatively smaller (<1-fold), but resulted in a significant increase in measured elastic modulus for the thinnest sections (10 μm) relative to the thicker (20 and 50 μm) sections. We also found that the measured elastic modulus depends modestly (again <1-fold), but significantly, on the magnitude of force applied, but only on thick (50 μm) and not thin (10 μm) tissue sections. Taken together these results demonstrate a dominant effect of indenter shape/radius on the measured elastic modulus of pulmonary arterial tissues, with lesser effects of tissue thickness and applied force. The results of this study highlight the importance of AFM parameter selection for accurate characterization of pulmonary arterial tissue mechanical properties, and allow for comparison of literature values for lung vessel tissue mechanical properties measured by AFM across a range of indenter and indentation parameters.
Collapse
|
22
|
Fan X, Zhu L, Wang K, Wang B, Wu Y, Xie W, Huang C, Chan BP, Du Y. Stiffness-Controlled Thermoresponsive Hydrogels for Cell Harvesting with Sustained Mechanical Memory. Adv Healthc Mater 2017; 6. [PMID: 28105774 DOI: 10.1002/adhm.201601152] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/23/2016] [Indexed: 01/17/2023]
Abstract
Most mechanobiological investigations focused on in situ mechanical regulation of cells on stiffness-controlled substrates with few downstream applications, as it is still challenging to harvest and expand mechanically primed cells by enzymatic digestion (e.g., trypsin) without interrupting cellular mechanical memory between passages. This study develops thermoresponsive hydrogels with controllable stiffness to generate mechanically primed cells with intact mechanical memory for augmented wound healing. No significant cellular property alteration of the fibroblasts primed on thermoresponsive hydrogels with varied stiffness has been observed through thermoresponsive harvesting. When reseeding the harvested cells for further evaluation, softer hydrogels are proven to better sustain the mechanical priming effects compared to rigid tissue culture plate, which indicates that both the stiffness-controlled substrate and thermoresponsive harvesting are required to sustain cellular mechanical memory between passages. Moreover, epigenetics analysis reveals that thermoresponsive harvesting could reduce the rearrangement and loss of chromatin proteins compared to that of trypsinization. In vivo wound healing using mechanically primed fibroblasts shows featured epithelium and sebaceous glands, which indicates augmented skin recovery compared with trypsinized fibroblasts. Thus, the thermoresponsive hydrogel-based cell harvesting system offers a powerful tool to investigate mechanobiology between cell passages and produces abundant cells with tailored mechanical priming properties for cell-based applications.
Collapse
Affiliation(s)
- Xingliang Fan
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
- Joint Center for Life Sciences; Tsinghua University-Peking University; Beijing 100084 China
| | - Lu Zhu
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
- Institute of Medical Equipment; Academy of Military Medical Sciences; Tianjin 300161 China
| | - Ke Wang
- Department of Chemistry; School of Science; Tsinghua University; Beijing 100084 China
| | - Bingjie Wang
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
- School of Life Science; Tsinghua University; Beijing 100084 China
| | - Yaozu Wu
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
| | - Wei Xie
- Joint Center for Life Sciences; Tsinghua University-Peking University; Beijing 100084 China
- School of Life Science; Tsinghua University; Beijing 100084 China
| | - Chengyu Huang
- Department of Plastic; Reconstructive and Aesthetic Surgery; Beijing Tsinghua Changgung Hospital; Tsinghua University; Beijing 102218 China
| | - Barbara Pui Chan
- Tissue Engineering Laboratory; Department of Mechanical Engineering; The University of Hong Kong; Pokfulam Road Hong Kong Special Administrative Region China
| | - Yanan Du
- Department of Biomedical Engineering; School of Medicine; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases; Tsinghua University; Beijing 100084 China
| |
Collapse
|
23
|
Hellewell AL, Rosini S, Adams JC. A Rapid, Scalable Method for the Isolation, Functional Study, and Analysis of Cell-derived Extracellular Matrix. J Vis Exp 2017. [PMID: 28117783 PMCID: PMC5351878 DOI: 10.3791/55051] [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] [Indexed: 12/15/2022] Open
Abstract
The extracellular matrix (ECM) is recognized as a diverse, dynamic, and complex environment that is involved in multiple cell-physiological and pathological processes. However, the isolation of ECM, from tissues or cell culture, is complicated by the insoluble and cross-linked nature of the assembled ECM and by the potential contamination of ECM extracts with cell surface and intracellular proteins. Here, we describe a method for use with cultured cells that is rapid and reliably removes cells to isolate a cell-derived ECM for downstream experimentation. Through use of this method, the isolated ECM and its components can be visualized by in situ immunofluorescence microscopy. The dynamics of specific ECM proteins can be tracked by tracing the deposition of a tagged protein using fluorescence microscopy, both before and after the removal of cells. Alternatively, the isolated ECM can be extracted for biochemical analysis, such as sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) and immunoblotting. At larger scales, a full proteomics analysis of the isolated ECM by mass spectrometry can be conducted. By conducting ECM isolation under sterile conditions, sterile ECM layers can be obtained for functional or phenotypic studies with any cell of interest. The method can be applied to any adherent cell type, is relatively easy to perform, and can be linked to a wide repertoire of experimental designs.
Collapse
|
24
|
Hoshiba T. Cultured cell-derived decellularized matrices: a review towards the next decade. J Mater Chem B 2017; 5:4322-4331. [DOI: 10.1039/c7tb00074j] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Summary of recent progress in cell-derived decellularized matrices preparation and application, with perspectives towards the next decade.
Collapse
Affiliation(s)
- T. Hoshiba
- Frontier Center for Organic Materials
- Yamagata University
- Yonezawa
- Japan
- Innovative Flex Course for Frontier Organic Materials Systems
| |
Collapse
|
25
|
Pizzute T, Zhang Y, He F, Pei M. Ascorbate-dependent impact on cell-derived matrix in modulation of stiffness and rejuvenation of infrapatellar fat derived stem cells toward chondrogenesis. Biomed Mater 2016; 11:045009. [PMID: 27508528 PMCID: PMC5004760 DOI: 10.1088/1748-6041/11/4/045009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Developing an in vitro microenvironment using cell-derived decellularized extracellular matrix (dECM) is a promising approach to efficiently expand adult stem cells for cartilage engineering and regeneration. Ascorbic acid serves as a critical stimulus for cells to synthesize collagens, which constitute the major component of dECM. In this study, we hypothesized that optimization of ascorbate treatment would maximize the rejuvenation effect of dECM on expanded stem cells from human infrapatellar fat pad in both proliferation and chondrogenic differentiation. In the duration regimen study, we found that dECM without L-ascorbic acid phosphate (AA) treatment, exhibiting lower stiffness measured by atomic force microscopy, yielded expanded cells with higher proliferation capacity but lower chondrogenic potential when compared to those with varied durations of AA treatment. dECM with 250 µM of AA treatment for 10 d had better rejuvenation in chondrogenic capacity if the deposited cells were from passage 2 rather than passage 5, despite no significant difference in matrix stiffness. In the dose regimen study, we found that dECMs deposited by varied concentrations of AA yielded expanded cells with higher proliferation capacity despite lower expression levels of stem cell related surface markers. Compared to cells expanded on tissue culture polystyrene, those on dECM exhibited greater chondrogenic potential, particularly for the dECMs with 50 µM and 250 µM of AA treatment. With the supplementation of ethyl-3,4-dihydroxybenzoate (EDHB), an inhibitor targeting procollagen synthesis, the dECM with 50 µM of AA treatment exhibited a dramatic decrease in the rejuvenation effect of expanded cell chondrogenic potential at both mRNA and protein levels despite no significant difference in matrix stiffness. Defined AA treatments during matrix preparation will benefit dECM-mediated stem cell engineering and future treatments for cartilage defects.
Collapse
Affiliation(s)
- Tyler Pizzute
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Ying Zhang
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV, USA
| | - Fan He
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Exercise Physiology, West Virginia University, Morgantown, WV, USA
- Mechanical and Aerospace Engineering, West Virginia University, Morgantown, WV, USA
- Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
26
|
Impaired von Willebrand factor adhesion and platelet response in thrombospondin-2 knockout mice. Blood 2016; 128:1642-50. [PMID: 27471233 DOI: 10.1182/blood-2016-03-702845] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 07/01/2016] [Indexed: 11/20/2022] Open
Abstract
Interactions between collagenous extracellular matrices and von Willebrand factor (VWF) are critical for hemostasis and thrombosis. In the present study, we investigated the contribution of an extracellular matrix (ECM) abnormality to the bleeding diathesis in thrombospondin-2 (TSP2) knockout (KO) mice. First, we performed adoptive bone marrow transplantation and observed that introduction of wild-type (WT) marrow into lethally irradiated TSP2 KO mice did not rescue the bleeding diathesis. However, platelets in transplanted mice displayed an inherent aggregation defect, which complicated interpretation. Second, we performed interposition of arterial segments denuded of endothelium. Denuded TSP2 KO arteries grafted into WT mice remained patent in vivo. In contrast, WT grafts underwent thrombosis and were completely occluded within 24 to 48 hours. The nonthrombogenic property of the TSP2 KO ECM was confirmed in vitro by exposing platelets to TSP2 KO dermal fibroblast (DF)-derived ECM. To further probe the effect of TSP2 deficiency, ECM production and deposition by WT and TSP2 KO DFs was analyzed via polymerase chain reaction, immunofluorescence, and scanning electron microscopy and showed similar patterns. In addition, atomic force microscopy (AFM) analysis of WT and TSP2 KO ECM did not reveal differences in stiffness. In contrast, reduced VWF accumulation on TSP2 KO ECM was observed when matrices were subjected to plasma under physiological flow. AFM utilizing VWF-coated 2-μm beads confirmed the weak binding to TSP2 KO ECM, providing a mechanistic explanation for the lack of thrombus formation. Therefore, our studies show that ECM assembly is critical for interaction of collagen with VWF and subsequent thrombogenic responses.
Collapse
|
27
|
Jorba I, Uriarte JJ, Campillo N, Farré R, Navajas D. Probing Micromechanical Properties of the Extracellular Matrix of Soft Tissues by Atomic Force Microscopy. J Cell Physiol 2016; 232:19-26. [PMID: 27163411 DOI: 10.1002/jcp.25420] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 05/09/2016] [Indexed: 01/11/2023]
Abstract
The extracellular matrix (ECM) determines 3D tissue architecture and provides structural support and chemical and mechanical cues to the cells. Atomic force microscopy (AFM) has unique capabilities to measure ECM mechanics at the scale at which cells probe the mechanical features of their microenvironment. Moreover, AFM measurements can be readily combined with bright field and fluorescence microscopy. Performing reliable mechanical measurements with AFM requires accurate calibration of the device and correct computation of the mechanical parameters. A suitable approach to isolate ECM mechanics from cell contribution is removing the cells by means of an effective decellularization process that preserves the composition, structure and mechanical properties of the ECM. AFM measurement of ECM micromechanics provides important insights into organ biofabrication, cell-matrix mechanical crosstalk and disease-induced tissue stiffness alterations. J. Cell. Physiol. 232: 19-26, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ignasi Jorba
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Juan J Uriarte
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain
| | - Noelia Campillo
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain.,CIBER de Enfermedades Respiratorias, Madrid, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Daniel Navajas
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain. .,Institute for Bioengineering of Catalonia (IBEC), Barcelona, Spain. .,CIBER de Enfermedades Respiratorias, Madrid, Spain.
| |
Collapse
|
28
|
Serbo JV, Kuo S, Lewis S, Lehmann M, Li J, Gracias DH, Romer LH. Patterning of Fibroblast and Matrix Anisotropy within 3D Confinement is Driven by the Cytoskeleton. Adv Healthc Mater 2016; 5:146-58. [PMID: 26033825 PMCID: PMC5817161 DOI: 10.1002/adhm.201500030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 05/12/2015] [Indexed: 12/16/2022]
Abstract
Effects of 3D confinement on cellular growth and matrix assembly are important in tissue engineering, developmental biology, and regenerative medicine. Polydimethylsiloxane wells with varying anisotropy are microfabicated using soft-lithography. Microcontact printing of bovine serum albumin is used to block cell adhesion to surfaces between wells. The orientations of fibroblast stress fibers, microtubules, and fibronectin fibrils are examined 1 day after cell seeding using laser scanning confocal microscopy, and anisotropy is quantified using a custom autocorrelation analysis. Actin, microtubules, and fibronectin exhibit higher anisotropy coefficients for cells grown in rectangular wells with aspect ratios of 1:4 and 1:8, as compared to those in wells with lower aspect ratios or in square wells. The effects of disabling individual cytoskeletal components on fibroblast responses to anisotropy are then tested by applying actin or microtubule polymerization inhibitors, Rho kinase inhibitor, or by siRNA-mediated knockdown of AXL or cofilin-1. Latrunculin A decreases cytoskeletal and matrix anisotropy, nocodazole ablates both, and Y27632 mutes cellular polarity while decreasing matrix anisotropy. AXL siRNA knockdown has little effect, as does siRNA knockdown of cofilin-1. These data identify several specific cytoskeletal strategies as targets for the manipulation of anisotropy in 3D tissue constructs.
Collapse
Affiliation(s)
- Janna V. Serbo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Scot Kuo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shawna Lewis
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Matthew Lehmann
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jiuru Li
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - David H. Gracias
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lewis H. Romer
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Generating and characterizing the mechanical properties of cell-derived matrices using atomic force microscopy. Methods 2015; 94:85-100. [PMID: 26439175 DOI: 10.1016/j.ymeth.2015.09.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/26/2015] [Accepted: 09/14/2015] [Indexed: 12/22/2022] Open
Abstract
Mechanical interaction between cells and their surrounding extracellular matrix (ECM) controls key processes such as proliferation, differentiation and motility. For many years, two-dimensional (2D) models were used to better understand the interactions between cells and their surrounding ECM. More recently, variation of the mechanical properties of tissues has been reported to play a major role in physiological and pathological scenarios such as cancer progression. The 3D architecture of the ECM finely tunes cellular behavior to perform physiologically relevant tasks. Technical limitations prevented scientists from obtaining accurate assessment of the mechanical properties of physiologically realistic matrices. There is therefore a need for combining the production of high-quality cell-derived 3D matrices (CDMs) and the characterization of their topographical and mechanical properties. Here, we describe methods that allow to accurately measure the young modulus of matrices produced by various cellular types. In the first part, we will describe and review several protocols for generating CDMs matrices from endothelial, epithelial, fibroblastic, muscle and mesenchymal stem cells. We will discuss tools allowing the characterization of the topographical details as well as of the protein content of such CDMs. In a second part, we will report the methodologies that can be used, based on atomic force microscopy, to accurately evaluate the stiffness properties of the CDMs through the quantification of their young modulus. Altogether, such methodologies allow characterizing the stiffness and topography of matrices deposited by the cells, which is key for the understanding of cellular behavior in physiological conditions.
Collapse
|
30
|
Revealing cytokine-induced changes in the extracellular matrix with secondary ion mass spectrometry. Acta Biomater 2015; 14:70-83. [PMID: 25523877 DOI: 10.1016/j.actbio.2014.12.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/22/2014] [Accepted: 12/08/2014] [Indexed: 11/23/2022]
Abstract
Cell-secreted matrices (CSMs), where extracellular matrix (ECM) deposited by monolayer cell cultures is decellularized, have been increasingly used to produce surfaces that may be reseeded with cells. Such surfaces are useful to help us understand cell-ECM interactions in a microenvironment closer to the in vivo situation than synthetic substrates with adsorbed proteins. We describe the production of CSMs from mouse primary osteoblasts (mPObs) exposed to cytokine challenge during matrix secretion, mimicking in vivo inflammatory environments. Time-of-flight secondary ion mass spectrometry data revealed that CSMs with cytokine challenge at day 7 or 12 of culture can be chemically distinguished from one another and from untreated CSM using multivariate analysis. Comparison of the differences with reference spectra from adsorbed protein mixtures points towards cytokine challenge resulting in a decrease in collagen content. This is supported by immunocytochemical and histological staining, demonstrating a 44% loss of collagen mass and a 32% loss in collagen I coverage. CSM surfaces demonstrate greater cell adhesion than adsorbed ECM proteins. When mPObs were reseeded onto cytokine-challenged CSMs they exhibited reduced adhesion and elongated morphology compared to untreated CSMs. Such changes may direct subsequent cell fate and function, and provide insights into pathological responses at sites of inflammation.
Collapse
|
31
|
Shkumatov A, Thompson M, Choi KM, Sicard D, Baek K, Kim DH, Tschumperlin DJ, Prakash YS, Kong H. Matrix stiffness-modulated proliferation and secretory function of the airway smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2015; 308:L1125-35. [PMID: 25724668 DOI: 10.1152/ajplung.00154.2014] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 02/27/2015] [Indexed: 01/02/2023] Open
Abstract
Multiple pulmonary conditions are characterized by an abnormal misbalance between various tissue components, for example, an increase in the fibrous connective tissue and loss/increase in extracellular matrix proteins (ECM). Such tissue remodeling may adversely impact physiological function of airway smooth muscle cells (ASMCs) responsible for contraction of airways and release of a variety of bioactive molecules. However, few efforts have been made to understand the potentially significant impact of tissue remodeling on ASMCs. Therefore, this study reports how ASMCs respond to a change in mechanical stiffness of a matrix, to which ASMCs adhere because mechanical stiffness of the remodeled airways is often different from the physiological stiffness. Accordingly, using atomic force microscopy (AFM) measurements, we found that the elastic modulus of the mouse bronchus has an arithmetic mean of 23.1 ± 14 kPa (SD) (median 18.6 kPa). By culturing ASMCs on collagen-conjugated polyacrylamide hydrogels with controlled elastic moduli, we found that gels designed to be softer than average airway tissue significantly increased cellular secretion of vascular endothelial growth factor (VEGF). Conversely, gels stiffer than average airways stimulated cell proliferation, while reducing VEGF secretion and agonist-induced calcium responses of ASMCs. These dependencies of cellular activities on elastic modulus of the gel were correlated with changes in the expression of integrin-β1 and integrin-linked kinase (ILK). Overall, the results of this study demonstrate that changes in matrix mechanics alter cell proliferation, calcium signaling, and proangiogenic functions in ASMCs.
Collapse
Affiliation(s)
- Artem Shkumatov
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | | | - Kyoung M Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Delphine Sicard
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Kwanghyun Baek
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Dong Hyun Kim
- Korea Institute of Industrial Technology, Ansan-si, South Korea
| | - Daniel J Tschumperlin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Y S Prakash
- Department of Anesthesiology, Mayo Clinic, Rochester, Minnesota
| | - Hyunjoon Kong
- Departments of Chemical and Biomolecular Engineering, Pathobiology, and Bioengineering, Institute of Genomic Biology, Univeristy of Illinois at Urbana-Champaign, Urbana, Illinois; and Deptartment of Chemical Engineering, Soongshil University, Seoul, Korea
| |
Collapse
|
32
|
Soucy PA, Hoh M, Heinz W, Hoh J, Romer L. Oriented matrix promotes directional tubulogenesis. Acta Biomater 2015; 11:264-73. [PMID: 25219769 PMCID: PMC4256113 DOI: 10.1016/j.actbio.2014.08.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 07/02/2014] [Accepted: 08/28/2014] [Indexed: 12/22/2022]
Abstract
Detailed control over the structural organization of scaffolds and engineered tissue constructs is a critical need in the quest to engineer functional tissues using biomaterials. This work presents a new approach to spatially direct endothelial tubulogenesis. Micropatterned fibronectin substrates were used to control lung fibroblast adhesion and growth and the subsequent deposition of fibroblast-derived matrix during culture. The fibroblast-derived matrix produced on the micropatterned substrates was tightly oriented by these patterns, with an average variation of only 8.5°. Further, regions of this oriented extracellular matrix provided directional control of developing endothelial tubes to within 10° of the original micropatterned substrate design. Endothelial cells seeded directly onto the micropatterned substrate did not form tubes. A metric for matrix anisotropy showed a relationship between the fibroblast-derived matrix and the endothelial tubes that were subsequently developed on the same micropatterns with a resulting aspect ratio over 1.5 for endothelial tubulogenesis. Micropatterns in "L" and "Y" shapes were used to direct endothelial tubes to turn and branch with the same level of precision. These data demonstrate that anisotropic fibroblast-derived matrices instruct the alignment and shape of endothelial tube networks, thereby introducing an approach that could be adapted for future design of microvascular implants featuring organ-specific natural matrix that patterns microvascular growth.
Collapse
Affiliation(s)
- Patricia A Soucy
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA
| | - Maria Hoh
- Intelligent Substrates, Inc., Sykesville, MD, USA
| | - Will Heinz
- Intelligent Substrates, Inc., Sykesville, MD, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jan Hoh
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lewis Romer
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Anesthesiology and Critical Care Medicine, Cell Biology, and Pediatrics, and the Center for Cell Dynamics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
33
|
Chang F, Lemmon CA, Nilaratanakul V, Rotter V, Romer L. Endothelial matrix assembly during capillary morphogenesis: insights from chimeric TagRFP-fibronectin matrix. J Histochem Cytochem 2014; 62:774-90. [PMID: 25063001 PMCID: PMC4209295 DOI: 10.1369/0022155414547419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/13/2014] [Indexed: 11/22/2022] Open
Abstract
Biologically relevant, three-dimensional extracellular matrix is an essential component of in vitro vasculogenesis models. WI-38 fibroblasts assemble a 3D matrix that induces endothelial tubulogenesis, but this model is challenged by fibroblast senescence and the inability to distinguish endothelial cell-derived matrix from matrix made by WI-38 fibroblasts. Matrices produced by hTERT-immortalized WI-38 recapitulated those produced by wild type fibroblasts. ECM fibrils were heavily populated by tenascin-C, fibronectin, and type VI collagen. Nearly half of the total type I collagen, but only a small fraction of the type IV collagen, were incorporated into ECM. Stable hTERT-WI-38 transfectants expressing TagRFP-fibronectin incorporated TagRFP into ~90% of the fibronectin in 3D matrices. TagRFP-fibronectin colocalized with tenascin-C and with type I collagen in a pattern that was similar to that seen in matrices from wild type WI-38. Human Umbilical Vein Endothelial Cells (HUVEC) formed 3D adhesions and tubes on WI38-hTERT-TagRFP-FN-derived matrices, and the TagRFP-fibronectin component of this new 3D human fibroblast matrix model facilitated the demonstration of concentrated membrane type 1 metalloprotease and new HUVEC FN and collagen type IV fibrils during EC tubulogenesis. These findings indicate that WI-38-hTERT- and WI-38-hTERT-TagRFP-FN-derived matrices provide platforms for the definition of new matrix assembly and remodeling events during vasculogenesis.
Collapse
Affiliation(s)
- Fumin Chang
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Christopher A Lemmon
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Voraphoj Nilaratanakul
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Varda Rotter
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| | - Lewis Romer
- Anesthesiology and Critical Care Medicine (FC, LR), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA (CAL)Cell Biology (LR), Johns Hopkins Medical Institutions, Baltimore, MDBiomedical Engineering (LR), Johns Hopkins Medical Institutions, Baltimore, MDPediatrics (LR), Johns Hopkins Medical Institutions, Baltimore, MDCenter for Cell Dynamics (LR), Johns Hopkins Medical Institutions, Baltimore, MDGraduate Program in Cellular and Molecular Medicine (VN), Johns Hopkins Medical Institutions, Baltimore, MDDepartment of Molecular and Cell Biology, The Weizmann Institute of Science, Rehovot, Israel (VR)
| |
Collapse
|
34
|
Herrmann D, Conway JRW, Vennin C, Magenau A, Hughes WE, Morton JP, Timpson P. Three-dimensional cancer models mimic cell-matrix interactions in the tumour microenvironment. Carcinogenesis 2014; 35:1671-9. [PMID: 24903340 DOI: 10.1093/carcin/bgu108] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Basic in vitro systems can be used to model and assess complex diseases, such as cancer. Recent advances in this field include the incorporation of multiple cell types and extracellular matrix proteins into three-dimensional (3D) models to recapitulate the structure, organization and functionality of live tissue in situ. Cells within such a 3D environment behave very differently from cells on two-dimensional (2D) substrates, as cell-matrix interactions trigger signalling pathways and cellular responses in 3D, which may not be observed in 2D. Thus, the use of 3D systems can be advantageous for the assessment of disease progression over 2D set-ups alone. Here, we highlight the current advantages and challenges of employing 3D systems in the study of cancer and provide an overview to guide the appropriate use of distinct models in cancer research.
Collapse
Affiliation(s)
- David Herrmann
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - James R W Conway
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Claire Vennin
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Astrid Magenau
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - William E Hughes
- Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and
| | - Jennifer P Morton
- The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| | - Paul Timpson
- Cancer Division, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia, Diabetes and Obesity Division, Garvan Institute of Medical Research, St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, NSW 2010, Sydney, Australia and The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
| |
Collapse
|
35
|
Abstract
Fibroblast migration is essential to normal wound healing and pathological matrix deposition in fibrosis. This review summarizes our understanding of how fibroblasts navigate 2D and 3D extracellular matrices, how this behavior is influenced by the architecture and mechanical properties of the matrix, and how migration is integrated with the other principle functions of fibroblasts, including matrix deposition, contraction, and degradation.
Collapse
Affiliation(s)
- Daniel J Tschumperlin
- Department of Environmental Health, Molecular and Integrative Physiological Sciences, Harvard School of Public Health, Boston, Massachusetts
| |
Collapse
|
36
|
Barreto-Ortiz SF, Zhang S, Davenport M, Fradkin J, Ginn B, Mao HQ, Gerecht S. A novel in vitro model for microvasculature reveals regulation of circumferential ECM organization by curvature. PLoS One 2013; 8:e81061. [PMID: 24278378 PMCID: PMC3836741 DOI: 10.1371/journal.pone.0081061] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 10/09/2013] [Indexed: 12/25/2022] Open
Abstract
In microvascular vessels, endothelial cells are aligned longitudinally whereas several components of the extracellular matrix (ECM) are organized circumferentially. While current three-dimensional (3D) in vitro models for microvasculature have allowed the study of ECM-regulated tubulogenesis, they have limited control over topographical cues presented by the ECM and impart a barrier for the high-resolution and dynamic study of multicellular and extracellular organization. Here we exploit a 3D fibrin microfiber scaffold to develop a novel in vitro model of the microvasculature that recapitulates endothelial alignment and ECM deposition in a setting that also allows the sequential co-culture of mural cells. We show that the microfibers' nanotopography induces longitudinal adhesion and alignment of endothelial colony-forming cells (ECFCs), and that these deposit circumferentially organized ECM. We found that ECM wrapping on the microfibers is independent of ECFCs' actin and microtubule organization, but it is dependent on the curvature of the microfiber. Microfibers with smaller diameters (100–400 µm) guided circumferential ECM deposition, whereas microfibers with larger diameters (450 µm) failed to support wrapping ECM. Finally, we demonstrate that vascular smooth muscle cells attached on ECFC-seeded microfibers, depositing collagen I and elastin. Collectively, we establish a novel in vitro model for the sequential control and study of microvasculature development and reveal the unprecedented role of the endothelium in organized ECM deposition regulated by the microfiber curvature.
Collapse
Affiliation(s)
- Sebastian F. Barreto-Ortiz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shuming Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Matthew Davenport
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jamie Fradkin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Brian Ginn
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
37
|
Kutys ML, Doyle AD, Yamada KM. Regulation of cell adhesion and migration by cell-derived matrices. Exp Cell Res 2013; 319:2434-9. [PMID: 23751565 DOI: 10.1016/j.yexcr.2013.05.030] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 01/16/2023]
Abstract
Three-dimensional in vitro extracellular matrix models provide a physiological alternative to regular two-dimensional cell culture, though they lack the full diversity of molecular composition and physical properties of whole-animal systems. Cell-derived matrices are extracellular matrices that are the product of matrix secretion and assembly by cells cultured at high density in vitro. After the removal of the cells that produced the matrix, an assembled matrix scaffold is left that closely mimics native stromal fiber organization and molecular content. Cell-derived matrices have been shown to impart in vivo-like responses to cells cultured in these matrices. In this review, we focus on mechanisms through which the distinct molecular and topographical composition of cell-derived matrices directs cellular behavior, specifically through regulation of cell-matrix adhesions and subsequent contributions to the process of cell migration.
Collapse
Affiliation(s)
- Matthew L Kutys
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research, United States.
| | | | | |
Collapse
|
38
|
Luque T, Melo E, Garreta E, Cortiella J, Nichols J, Farré R, Navajas D. Local micromechanical properties of decellularized lung scaffolds measured with atomic force microscopy. Acta Biomater 2013; 9:6852-9. [PMID: 23470549 DOI: 10.1016/j.actbio.2013.02.044] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 02/21/2013] [Accepted: 02/26/2013] [Indexed: 01/13/2023]
Abstract
Bioartificial lungs re-engineered from decellularized organ scaffolds are a promising alternative to lung transplantation. Critical features for improving scaffold repopulation depend on the mechanical properties of the cell microenvironment. However, the mechanics of the lung extracellular matrix (ECM) is poorly defined. The local mechanical properties of the ECM were measured in different regions of decellularized rat lung scaffolds with atomic force microscopy. Lungs excised from rats (n=11) were decellularized with sodium dodecyl sulfate (SDS) and cut into ~7μm thick slices. The complex elastic modulus (G(∗)) of lung ECM was measured over a frequency band ranging from 0.1 to 11.45Hz. Measurements were taken in alveolar wall segments, alveolar wall junctions and pleural regions. The storage modulus (G', real part of G(∗)) of alveolar ECM was ~6kPa, showing small changes between wall segments and junctions. Pleural regions were threefold stiffer than alveolar walls. G' of alveolar walls and pleura increased with frequency as a weak power law with exponent 0.05. The loss modulus (G″, imaginary part of G(∗)) was 10-fold lower and showed a frequency dependence similar to that of G' at low frequencies (0.1-1Hz), but increased more markedly at higher frequencies. Local differences in mechanical properties and topology of the parenchymal site could be relevant mechanical cues for regulating the spatial distribution, differentiation and function of lung cells.
Collapse
|
39
|
Roth M, Zhong J, Zumkeller C, S'ng CT, Goulet S, Tamm M. The role of IgE-receptors in IgE-dependent airway smooth muscle cell remodelling. PLoS One 2013; 8:e56015. [PMID: 23457493 PMCID: PMC3573085 DOI: 10.1371/journal.pone.0056015] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 01/09/2013] [Indexed: 02/07/2023] Open
Abstract
Background In allergic asthma, IgE increases airway remodelling but the mechanism is incompletely understood. Airway remodelling consists of two independent events increased cell numbers and enhanced extracellular matrix deposition, and the mechanism by which IgE up-regulates cell proliferation and extracellular matrix deposition by human airway smooth muscle cells in asthma is unclear. Objective Characterise the role of the two IgE receptors and associated signalling cascades in airway smooth muscle cell remodelling. Methods Primary human airway smooth muscle cells (8 asthmatics, 8 non-asthmatics) were stimulated with human purified antibody-activated IgE. Proliferation was determined by direct cell counts. Total collagen deposition was determined by Sircol; collagen species deposition by ELISA. IgE receptors were silenced by siRNA and mitogen activated protein kinase (MAPK) signalling was blocked by chemical inhibitors. Results IgE dose-dependently increased extracellular matrix and collagen deposition by airway smooth muscle cells as well as their proliferation. Specifically in cells of asthma patients IgE increased the deposition of collagen-type-I, -III, –VII and fibronectin, but did not affect the deposition of collagens type-IV. IgE stimulated collagen type-I and type-VII deposition through IgE receptor-I and Erk1/2 MAPK. Proliferation and deposition of collagens type-III and fibronectin involved both IgE receptors as well as Erk1/2 and p38 MAPK. Pre-incubation (30 minutes) with Omalizumab prevented all remodelling effects completely. We observed no changes in gelatinase activity or their inhibitors. Conclusion & Clincal Relevance Our study provides the molecular biological mechanism by which IgE increases airway remodelling in asthma through increased airway smooth muscle cell proliferation and deposition of pro-inflammatory collagens and fibronectin. Blocking IgE action prevents several aspects of airway smooth muscle cell remodelling. Our findings may explain the recently described reduction of airway wall thickness in severe asthma patients treated with humanised anti-IgE antibodies.
Collapse
Affiliation(s)
- Michael Roth
- Pulmonary Cell Research, Department Biomedicine, University of Basel, Basel, Switzerland.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Contractile myofibroblasts are responsible for the irreversible alterations of the lung parenchyma that hallmark pulmonary fibrosis. In response to lung injury, a variety of different precursor cells can become activated to develop myofibroblast features, most notably formation of stress fibers and expression of α-smooth muscle actin. Starting as an acute and beneficial repair process, myofibroblast secretion of collagen and contraction frequently becomes excessive and persists. The result is accumulation of stiff scar tissue that obstructs and ultimately destroys lung function. In addition to being a consequence of myofibroblast activities, the stiffened tissue is also a major promoter of the myofibroblast. The mechanical properties of scarred lung and fibrotic foci promote myofibroblast contraction and differentiation. One essential element in this detrimental feed-forward loop is the mechanical activation of the profibrotic growth factor transforming growth factor-β1 from stores in the extracellular matrix. Interfering with myofibroblast contraction and integrin-mediated force transmission to latent transforming growth factor-β1 and matrix proteins are here presented as possible therapeutic strategies to halt fibrosis.
Collapse
|
41
|
Atari M, Caballé-Serrano J, Gil-Recio C, Giner-Delgado C, Martínez-Sarrà E, García-Fernández DA, Barajas M, Hernández-Alfaro F, Ferrés-Padró E, Giner-Tarrida L. The enhancement of osteogenesis through the use of dental pulp pluripotent stem cells in 3D. Bone 2012; 50:930-41. [PMID: 22270057 DOI: 10.1016/j.bone.2012.01.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 12/30/2011] [Accepted: 01/02/2012] [Indexed: 01/09/2023]
Abstract
The potential for osteogenic differentiation of dental pulp mesenchymal stem cells (DPMSCs) in vitro and in vivo has been well documented in a variety of studies. Previously, we obtained a population of cells from human dental pulp called dental pulp pluripotent stem cells (DPPSCs) that could differentiate into mesodermal, ectodermal and endodermal progenies. We compared the osteogenic capacity of DPPSCs and DPMSCs that had been isolated from the same donors (N=5) and cultivated in the same osteogenic medium in 3D (three dimensions) Cell Carrier glass scaffolds. We also compared the architecture of bone-like tissue obtained from DPPSCs and human maxillary bone tissue. Differentiation was evaluated by scanning electron microscopy, whereas the expression of bone markers such as ALP, Osteocalcin, COLL1 and Osteonectin was investigated by quantitative real time polymerase chain reaction (qRT-PCR). We also used calcium quantification, Alizarin red staining and alkaline phosphatase (ALP) activity to compare the two cell types. New bone tissue formed by DPPSCs was in perfect continuity with the trabecular host bone structure, and the restored bone network demonstrated high interconnectivity. Significant differences between DPPSCs and DPMSCs were observed for the expression of bone markers, calcium deposition and ALP activity during osteogenic differentiation; these criteria were higher for DPPSCs than DPMSCs. Both DPPSCs and differentiated tissue showed normal chromosomal dosage after being cultured in vitro and analysed using short-chromosome genomic hybridisation (short-CGH). This study demonstrates the stability and potential for the use of DPPSCs in bone tissue engineering applications.
Collapse
Affiliation(s)
- M Atari
- Laboratory for Regenerative Medicine, Department of Oral and Maxillofacial Surgery, College of Dentistry, Universitat Internacional de Catalunya, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mishra DK, Thrall MJ, Baird BN, Ott HC, Blackmon SH, Kurie JM, Kim MP. Human lung cancer cells grown on acellular rat lung matrix create perfusable tumor nodules. Ann Thorac Surg 2012; 93:1075-81. [PMID: 22385822 PMCID: PMC4512949 DOI: 10.1016/j.athoracsur.2012.01.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 01/03/2012] [Accepted: 01/06/2012] [Indexed: 11/30/2022]
Abstract
BACKGROUND Extracellular matrix allows lung cancer to form its shape and grow. Recent studies on organ reengineering for orthotopic transplantation have provided a new avenue for isolating purified native matrix to use for growing cells. Whether human lung cancer cells grown in a decellularized rat lung matrix would create perfusable human lung cancer nodules was tested. METHODS Rat lungs were harvested and native cells were removed using sodium dodecyl sulfate and Triton X-100 in a decellularization chamber to create a decellularized rat lung matrix. Human A549, H460, or H1299 lung cancer cells were placed into the decellularized rat lung matrix and grown in a customized bioreactor with perfusion of oxygenated media for 7 to 14 days. RESULTS Decellularized rat lung matrix showed preservation of matrix architecture devoid of all rat cells. All three human lung cancer cell lines grown in the bioreactor developed tumor nodules with intact vasculature. Moreover, the lung cancer cells developed a pattern of growth similar to the original human lung cancer. CONCLUSIONS Overall, this study shows that human lung cancer cells form perfusable tumor nodules in a customized bioreactor on a decellularized rat lung matrix created by a customized decellularization chamber. The lung cancer cells grown in the matrix had features similar to the original human lung cancer. This ex vivo model can be used potentially to gain a deeper understanding of the biologic processes involved in human lung cancer.
Collapse
Affiliation(s)
- Dhruva K Mishra
- Department of Surgery, The Methodist Hospital Research Institute, Houston, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Balestrini JL, Chaudhry S, Sarrazy V, Koehler A, Hinz B. The mechanical memory of lung myofibroblasts. Integr Biol (Camb) 2012; 4:410-21. [PMID: 22410748 DOI: 10.1039/c2ib00149g] [Citation(s) in RCA: 259] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Fibroblasts differentiate into the highly synthetic and contractile myofibroblast phenotype when exposed to substrates with an elastic modulus corresponding to pathologically stiff fibrotic tissue. Cellular responses to changes in substrate stiffness are typically analyzed after hours or days, which does not enable the monitoring of myofibroblast persistence, a hallmark of fibrosis. To determine long-lasting effects on the fibrotic behavior of lung fibroblasts, we followed a novel approach of explanting and repeatedly passaging fibroblasts on silicone substrates with stiffness representing various states of lung health. Fibrotic activity was determined by assaying for myofibroblast proliferation, cell contractility, expression of α-smooth muscle actin, extracellular matrix and active TGFβ1. As predicted, myofibroblast activity was low on healthy soft substrates and increased with increasing substrate stiffness. However, explanting and mechanically priming lung fibroblasts for 3 weeks on pathologically stiff substrates resulted in sustained myofibroblast activity even after the cells were returned to healthy soft cultures for 2 weeks. Such primed cells retained higher fibrotic activity than cells that had been exclusively cultured on soft substrates, and were not statistically different from cells continuously passaged on stiff surfaces. Inversely, priming lung fibroblasts for 3 weeks on soft substrates partially protected from myofibroblast activation after the shift to stiff substrates. Hence, mechano-sensed information relating to physical conditions of the local cellular environment could permanently induce fibrotic behavior of lung fibroblasts. This priming effect has important implications for the progression and persistence of aggressive fibrotic diseases such as idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Jenna L Balestrini
- Laboratory of Tissue Repair and Regeneration, Matrix Dynamics Group, Faculty of Dentistry, University of Toronto, Fitzgerald Building, Room 234, 150 College Street, Toronto, ON M5S3E2, Canada
| | | | | | | | | |
Collapse
|