1
|
Metzcar J, Duggan BS, Fischer B, Murphy M, Heiland R, Macklin P. A Simple Framework for Agent-Based Modeling with Extracellular Matrix. Bull Math Biol 2025; 87:43. [PMID: 39937344 PMCID: PMC11821717 DOI: 10.1007/s11538-024-01408-8] [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: 02/02/2023] [Accepted: 12/21/2024] [Indexed: 02/13/2025]
Abstract
Extracellular matrix (ECM) is a key component of the cellular microenvironment and critical in multiple disease and developmental processes. Representing ECM and cell-ECM interactions is a challenging multiscale problem as they span molecular-level details to tissue-level dynamics. While several computational frameworks exist for ECM modeling, they often focus on very detailed modeling of individual ECM fibers or represent only a single aspect of the ECM. Using the PhysiCell agent-based modeling platform, we developed a framework of intermediate detail with the ability to capture bidirectional cell-ECM interactions. We represent a small region of ECM, an ECM element, with three variables describing its local microstructure: anisotropy, density, and overall fiber orientation. To spatially model the ECM, we use an array of ECM elements. Cells remodel local ECM microstructure and in turn, local microstructure impacts cellular motility. We demonstrate the utility of this framework and reusability of its core cell-ECM interaction model through examples in cellular invasion, wound healing, basement membrane degradation, and leader-follower collective migration. Despite the relative simplicity of the framework, it is able to capture a broad range of cell-ECM interactions of interest to the modeling community. Furthermore, variables representing the ECM microstructure are accessible through simple programming interfaces. This allows them to impact cell behaviors, such as proliferation and death, without requiring custom code for each interaction, particularly through PhysiCell's modeling grammar, enabling rapid modeling of a diverse range of cell-matrix biology. We make this framework available as a free and open source software package at https://github.com/PhysiCell-Models/collective-invasion .
Collapse
Affiliation(s)
- John Metzcar
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
- Informatics, Indiana University, 901 E. Tenth Street, Bloomington, IN, 47408, USA
| | - Ben S Duggan
- Computer Science, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Brandon Fischer
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Matthew Murphy
- Informatics, Indiana University, 901 E. Tenth Street, Bloomington, IN, 47408, USA
| | - Randy Heiland
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA
| | - Paul Macklin
- Intelligent Systems Engineering, Indiana University, 700 N. Woodlawn, Bloomington, IN, 47408, USA.
| |
Collapse
|
2
|
Xiao H, Sylla K, Gong X, Wilkowski B, Rossello-Martinez A, Jordan SN, Mintah EY, Zheng A, Sun H, Herzog EL, Mak M. Proteolysis and Contractility Regulate Tissue Opening and Wound Healing by Lung Fibroblasts in 3D Microenvironments. Adv Healthc Mater 2024; 13:e2400941. [PMID: 38967294 PMCID: PMC11617280 DOI: 10.1002/adhm.202400941] [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: 03/12/2024] [Revised: 05/17/2024] [Indexed: 07/06/2024]
Abstract
Damage and repair are recurring processes in tissues, with fibroblasts playing key roles by remodeling extracellular matrices (ECM) through protein synthesis, proteolysis, and cell contractility. Dysregulation of fibroblasts can lead to fibrosis and tissue damage, as seen in idiopathic pulmonary fibrosis (IPF). In advanced IPF, tissue damage manifests as honeycombing, or voids in the lungs. This study explores how transforming growth factor-beta (TGF-β), a crucial factor in IPF, induces lung fibroblast spheroids to create voids in reconstituted collagen through proteolysis and cell contractility, a process we termed as hole formation. These voids reduce when proteases are blocked. Spheroids mimic fibroblast foci observed in IPF. Results indicate that cell contractility mediates tissue opening by stretching fractures in the collagen meshwork. Matrix metalloproteinases (MMPs), including MMP1 and MT1-MMP, are essential for hole formation, with invadopodia playing a significant role. Blocking MMPs reduces hole size and promotes wound healing. This study shows how TGF-β induces excessive tissue destruction and how blocking proteolysis can reverse damage, offering insights into IPF pathology and potential therapeutic interventions.
Collapse
Affiliation(s)
- Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Kadidia Sylla
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Brendan Wilkowski
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | | | - Seyma Nayir Jordan
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Emmanuel Y Mintah
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Allen Zheng
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| | - Huanxing Sun
- Department of Medicine (Pulmonary, Critical Care and Sleep), Yale School of Medicine, New Haven, CT, 06510, USA
| | - Erica L Herzog
- Department of Medicine (Pulmonary, Critical Care and Sleep), Yale School of Medicine, New Haven, CT, 06510, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
| |
Collapse
|
3
|
Alshehri AM, Wilson OC. Biomimetic Hydrogel Strategies for Cancer Therapy. Gels 2024; 10:437. [PMID: 39057460 PMCID: PMC11275631 DOI: 10.3390/gels10070437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Recent developments in biomimetic hydrogel research have expanded the scope of biomedical technologies that can be used to model, diagnose, and treat a wide range of medical conditions. Cancer presents one of the most intractable challenges in this arena due to the surreptitious mechanisms that it employs to evade detection and treatment. In order to address these challenges, biomimetic design principles can be adapted to beat cancer at its own game. Biomimetic design strategies are inspired by natural biological systems and offer promising opportunities for developing life-changing methods to model, detect, diagnose, treat, and cure various types of static and metastatic cancers. In particular, focusing on the cellular and subcellular phenomena that serve as fundamental drivers for the peculiar behavioral traits of cancer can provide rich insights into eradicating cancer in all of its manifestations. This review highlights promising developments in biomimetic nanocomposite hydrogels that contribute to cancer therapies via enhanced drug delivery strategies and modeling cancer mechanobiology phenomena in relation to metastasis and synergistic sensing systems. Creative efforts to amplify biomimetic design research to advance the development of more effective cancer therapies will be discussed in alignment with international collaborative goals to cure cancer.
Collapse
Affiliation(s)
- Awatef M. Alshehri
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
- Department of Nanomedicine, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdelaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia;
| | - Otto C. Wilson
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
4
|
Wang W, Zanotelli MR, Sabo LN, Fabiano ED, Goldfield NM, Le C, Techasiriwan EP, Lopez S, Berestesky ED, Reinhart-King CA. Collagen density regulates tip-stalk cell rearrangement during angiogenesis via cellular bioenergetics. APL Bioeng 2024; 8:026120. [PMID: 38872716 PMCID: PMC11170328 DOI: 10.1063/5.0195249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 05/07/2024] [Indexed: 06/15/2024] Open
Abstract
Tumor vasculature plays a crucial role in tumor progression, affecting nutrition and oxygen transportation as well as the efficiency of drug delivery. While targeting pro-angiogenic growth factors has been a significant focus for treating tumor angiogenesis, recent studies indicate that metabolism also plays a role in regulating endothelial cell behavior. Like cancer cells, tumor endothelial cells undergo metabolic changes that regulate rearrangement for tip cell position during angiogenesis. Our previous studies have shown that altered mechanical properties of the collagen matrix regulate angiogenesis and can promote a tumor vasculature phenotype. Here, we examine the effect of collagen density on endothelial cell tip-stalk cell rearrangement and cellular energetics during angiogenic sprouting. We find that increased collagen density leads to an elevated energy state and an increased rate of tip-stalk cell switching, which is correlated with the energy state of the cells. Tip cells exhibit higher glucose uptake than stalk cells, and inhibition of glucose uptake revealed that invading sprouts rely on glucose to meet elevated energy requirements for invasion in dense matrices. This work helps to elucidate the complex interplay between the mechanical microenvironment and the endothelial cell metabolic status during angiogenesis, which could have important implications for developing new anti-cancer therapies.
Collapse
Affiliation(s)
- Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | | | - Lindsey N. Sabo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Emily D. Fabiano
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Natalie M. Goldfield
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Chloe Le
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Elle P. Techasiriwan
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Santiago Lopez
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Emily D. Berestesky
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | | |
Collapse
|
5
|
Leartprapun N, Zeng Z, Hajjarian Z, Bossuyt V, Nadkarni SK. Laser speckle rheological microscopy reveals wideband viscoelastic spectra of biological tissues. SCIENCE ADVANCES 2024; 10:eadl1586. [PMID: 38718128 PMCID: PMC11078189 DOI: 10.1126/sciadv.adl1586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
Viscoelastic transformation of tissue drives aberrant cellular functions and is an early biomarker of disease pathogenesis. Tissues scale a range of viscoelastic moduli, from biofluids to bone. Moreover, viscoelastic behavior is governed by the frequency at which tissue is probed, yielding distinct viscous and elastic responses modulated over a wide frequency band. Existing tools do not quantify wideband viscoelastic spectra in tissues, leaving a vast knowledge gap. We present wideband laser speckle rheological microscopy (WB-SHEAR) that reveals elastic and viscous response over sub-megahertz frequencies previously not investigated in tissue. WB-SHEAR uses an optical, noncontact approach to quantify wideband viscoelastic spectra in specimens spanning a range of moduli from low-viscosity fibrin to highly elastic bone. Via laser scanning, micromechanical imaging is enabled to access wideband viscoelastic spectra in heterogeneous tumor specimens with high spatial resolution (25 micrometers). The ability to interrogate the viscoelastic landscape of diverse biospecimens could transform our understanding of mechanobiological processes in various diseases.
Collapse
Affiliation(s)
- Nichaluk Leartprapun
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Ziqian Zeng
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Zeinab Hajjarian
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Veerle Bossuyt
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Seemantini K. Nadkarni
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
6
|
Tan ML, Jenkins-Johnston N, Huang S, Schutrum B, Vadhin S, Adhikari A, Williams RM, Zipfel WR, Lammerding J, Varner JD, Fischbach C. Endothelial cells metabolically regulate breast cancer invasion toward a microvessel. APL Bioeng 2023; 7:046116. [PMID: 38058993 PMCID: PMC10697723 DOI: 10.1063/5.0171109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/06/2023] [Indexed: 12/08/2023] Open
Abstract
Breast cancer metastasis is initiated by invasion of tumor cells into the collagen type I-rich stroma to reach adjacent blood vessels. Prior work has identified that metabolic plasticity is a key requirement of tumor cell invasion into collagen. However, it remains largely unclear how blood vessels affect this relationship. Here, we developed a microfluidic platform to analyze how tumor cells invade collagen in the presence and absence of a microvascular channel. We demonstrate that endothelial cells secrete pro-migratory factors that direct tumor cell invasion toward the microvessel. Analysis of tumor cell metabolism using metabolic imaging, metabolomics, and computational flux balance analysis revealed that these changes are accompanied by increased rates of glycolysis and oxygen consumption caused by broad alterations of glucose metabolism. Indeed, restricting glucose availability decreased endothelial cell-induced tumor cell invasion. Our results suggest that endothelial cells promote tumor invasion into the stroma due, in part, to reprogramming tumor cell metabolism.
Collapse
Affiliation(s)
- Matthew L. Tan
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Niaa Jenkins-Johnston
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Sarah Huang
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Brittany Schutrum
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Sandra Vadhin
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Abhinav Adhikari
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Rebecca M. Williams
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Warren R. Zipfel
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Jeffrey D. Varner
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, USA
| | | |
Collapse
|
7
|
Wang T, Huang Q, Rao Z, Liu F, Su X, Zhai X, Ma J, Liang Y, Quan D, Liao G, Bai Y, Zhang S. Injectable decellularized extracellular matrix hydrogel promotes salivary gland regeneration via endogenous stem cell recruitment and suppression of fibrogenesis. Acta Biomater 2023; 169:256-272. [PMID: 37557943 DOI: 10.1016/j.actbio.2023.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/26/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023]
Abstract
Saliva is key to the maintenance of oral homeostasis. However, several forms of salivary gland (SG) disorders, followed by hyposalivation, often result in dental caries, oral infection, and decreased taste, which dramatically affect the quality of patient's life. Functional biomaterials hold great potential for tissue regeneration in damaged or dysfunctional SGs and maintaining the good health of oral cavity. Herein, we prepared an injectable hydrogel derived from decellularized porcine submandibular glands (pDSG-gel), the material and biological properties of the hydrogel were systematically investigated. First, good biocompatibility and bioactivities of the pDSG-gel were validated in 2D and 3D cultures of primary submandibular gland mesenchymal stem cells (SGMSCs). Especially, the pDSG-gel effectively facilitated SGMSCs migration and recruitment through the activation of PI3K/AKT signaling pathway, suggested by transcriptomic analysis and immunoblotting. Furthermore, proteomic analysis of the pDSG revealed that many extracellular matrix components and secreted factors were preserved, which may contribute to stem cell homing. The recruitment of endogenous SG cells was confirmed in vivo, upon in situ injection of the pDSG-gel into the defective SGs in rats. Acinar and ductal-like structures were evident in the injury sites after pDSG-gel treatment, suggesting the reconstruction of functional SG units. Meanwhile, histological characterizations showed that the administration of the pDSG-gel also significantly suppressed fibrogenesis within the injured SG tissues. Taken together, this tissue-specific hydrogel provides a pro-regenerative microenvironment for endogenous SG regeneration and holds great promise as a powerful and bioactive material for future treatments of SG diseases. STATEMENT OF SIGNIFICANCE: Decellularized extracellular matrix (dECM) has been acknowledged as one of the most promising biomaterials that recapitalizes the microenvironment in native tissues. Hydrogel derived from the dECM allows in situ administration for tissue repair. Herein, a tissue-specific dECM hydrogel derived from porcine salivary glands (pDSG-gel) was successfully prepared and developed for functional reconstruction of defective salivary gland (SG) tissues. The pDSG-gel effectively accelerated endogenous SG stem cells migration and their recruitment for acinar- and ductal-like regeneration, which was attributed to the activation of PI3K/AKT signaling pathway. Additionally, the introduction of the pDSG-gel resulted in highly suppressed fibrogenesis in the defective tissues. These outcomes indicated that the pDSG-gel holds great potential in clinical translation toward SG regeneration through cell-free treatments.
Collapse
Affiliation(s)
- Tao Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Qiting Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Zilong Rao
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, PCFM Lab, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Fan Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Xinyun Su
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Xuefan Zhai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Jingxin Ma
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Yujie Liang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
| | - Daping Quan
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, PCFM Lab, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guiqing Liao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China.
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, PCFM Lab, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Sien Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China.
| |
Collapse
|
8
|
Leartprapun N, Zeng Z, Hajjarian Z, Bossuyt V, Nadkarni SK. Speckle rheological spectroscopy reveals wideband viscoelastic spectra of biological tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544037. [PMID: 37333220 PMCID: PMC10274797 DOI: 10.1101/2023.06.08.544037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Mechanical transformation of tissue is not merely a symptom but a decisive driver in pathological processes. Comprising intricate network of cells, fibrillar proteins, and interstitial fluid, tissues exhibit distinct solid-(elastic) and liquid-like (viscous) behaviours that span a wide band of frequencies. Yet, characterization of wideband viscoelastic behaviour in whole tissue has not been investigated, leaving a vast knowledge gap in the higher frequency range that is linked to fundamental intracellular processes and microstructural dynamics. Here, we present wideband Speckle rHEologicAl spectRoScopy (SHEARS) to address this need. We demonstrate, for the first time, analysis of frequency-dependent elastic and viscous moduli up to the sub-MHz regime in biomimetic scaffolds and tissue specimens of blood clots, breast tumours, and bone. By capturing previously inaccessible viscoelastic behaviour across the wide frequency spectrum, our approach provides distinct and comprehensive mechanical signatures of tissues that may provide new mechanobiological insights and inform novel disease prognostication.
Collapse
Affiliation(s)
- Nichaluk Leartprapun
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Ziqian Zeng
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Zeinab Hajjarian
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Veerle Bossuyt
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Seemantini K. Nadkarni
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
9
|
Nguyen RY, Cabral AT, Rossello-Martinez A, Zulli A, Gong X, Zhang Q, Yan J, Mak M. Tunable Mesoscopic Collagen Island Architectures Modulate Stem Cell Behavior. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2207882. [PMID: 36895051 PMCID: PMC10166061 DOI: 10.1002/adma.202207882] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/22/2022] [Indexed: 05/10/2023]
Abstract
The extracellular matrix is the biophysical environment that scaffolds mammalian cells in the body. The main constituent is collagen. In physiological tissues, collagen network topology is diverse with complex mesoscopic features. While studies have explored the roles of collagen density and stiffness, the impact of complex architectures remains not well-understood. Developing in vitro systems that recapitulate these diverse collagen architectures is critical for understanding physiologically relevant cell behaviors. Here, methods are developed to induce the formation of heterogeneous mesoscopic architectures, referred to as collagen islands, in collagen hydrogels. These island-containing gels have highly tunable inclusions and mechanical properties. Although these gels are globally soft, there is regional enrichment in the collagen concentration at the cell-scale. Collagen-island architectures are utilized to study mesenchymal stem cell behavior, and it is demonstrated that cell migration and osteogenic differentiation are altered. Finally, induced pluripotent stem cells are cultured in island-containing gels, and it is shown that the architecture is sufficient to induce mesodermal differentiation. Overall, this work highlights complex mesoscopic tissue architectures as bioactive cues in regulating cell behavior and presents a novel collagen-based hydrogel that captures these features for tissue engineering applications.
Collapse
Affiliation(s)
- Ryan Y. Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Aidan T. Cabral
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | | | - Alessandro Zulli
- Department of Chemical and Environmental Engineering, Yale University, New Haven, CT, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Qiuting Zhang
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Jing Yan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
10
|
3D collagen migration patterns reveal a SMAD3-dependent and TGF-β1-independent mechanism of recruitment for tumour-associated fibroblasts in lung adenocarcinoma. Br J Cancer 2023; 128:967-981. [PMID: 36572730 PMCID: PMC10006167 DOI: 10.1038/s41416-022-02093-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 11/19/2022] [Accepted: 11/25/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The TGF-β1 transcription factor SMAD3 is epigenetically repressed in tumour-associated fibroblasts (TAFs) from lung squamous cell carcinoma (SCC) but not adenocarcinoma (ADC) patients, which elicits a compensatory increase in SMAD2 that renders SCC-TAFs less fibrotic. Here we examined the effects of altered SMAD2/3 in fibroblast migration and its impact on the desmoplastic stroma formation in lung cancer. METHODS We used a microfluidic device to examine descriptors of early protrusions and subsequent migration in 3D collagen gels upon knocking down SMAD2 or SMAD3 by shRNA in control fibroblasts and TAFs. RESULTS High SMAD3 conditions as in shSMAD2 fibroblasts and ADC-TAFs exhibited a migratory advantage in terms of protrusions (fewer and longer) and migration (faster and more directional) selectively without TGF-β1 along with Erk1/2 hyperactivation. This enhanced migration was abrogated by TGF-β1 as well as low glucose medium and the MEK inhibitor Trametinib. In contrast, high SMAD2 fibroblasts were poorly responsive to TGF-β1, high glucose and Trametinib, exhibiting impaired migration in all conditions. CONCLUSIONS The basal migration advantage of high SMAD3 fibroblasts provides a straightforward mechanism underlying the larger accumulation of TAFs previously reported in ADC compared to SCC. Moreover, our results encourage using MEK inhibitors in ADC-TAFs but not SCC-TAFs.
Collapse
|
11
|
Edwin PERG, Kumar S, Roy S, Roy B, Bajpai SK. Anisotropic 3D confinement of MCF-7 cells induces directed cell-migration and viscoelastic anisotropy of cell-membrane. Phys Biol 2023; 20. [DOI: 10.1088/1478-3975/ac9bc1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022]
Abstract
Abstract
Tumor-associated collagen signature-3 (TACS-3) is a prognostic indicator for breast cancer survival. It is characterized by highly organized, parallel bundles of collagen fibers oriented perpendicular to the tumor boundary, serving as directional, confining channels for cancer cell invasion. Here we design a TACS-3-mimetic anisotropic, confined collagen I matrix and examine the relation between anisotropy of matrix, directed cellular migration, and anisotropy of cell membrane-the first direct contact between TACS-3 and cell-using Michigan Cancer Foundation-7 (MCF-7) cells as cancer-model. Using unidirectional freezing, we generated ∼50 μm-wide channels filled with collagen I. Optical tweezer (OT) microrheology shows that anisotropic confinement increases collagen viscoelasticity by two orders of magnitude, and the elastic modulus is significantly greater along the direction of anisotropic confinement compared to that along the orthogonal direction, thus establishing matrix anisotropy. Furthermore, MCF-7 cells embedded in anisotropic collagen I, exhibit directionality in cellular morphology and migration. Finally, using customized OT to trap polystyrene probes bound to cell-membrane (and not to ECM) of either free cells or cells under anisotropic confinement, we quantified the effect of matrix anisotropy on membrane viscoelasticity, both in-plane and out-of-plane, vis-à-vis the membrane. Both bulk and viscous modulus of cell-membrane of MCF-7 cells exhibit significant anisotropy under anisotropic confinement. Moreover, the cell membrane of MCF-7 cells under anisotropic confinement is significantly softer (both in-plane and out-of-plane moduli) despite their local environment being five times stiffer than free cells. In order to test if the coupling between anisotropy of extracellular matrix and anisotropy of cell-membrane is regulated by cell-cytoskeleton, actin cytoskeleton was depolymerized for both free and confined cells. Results show that cell membrane viscoelasticity of confined MCF-7 cells is unaffected by actin de-polymerization, in contrast to free cells. Together, these findings suggest that anisotropy of ECM induces directed migration and correlates with anisotropy of cell-membrane viscoelasticity of the MCF-7 cells in an actin-independent manner.
Collapse
|
12
|
Merino-Casallo F, Gomez-Benito MJ, Hervas-Raluy S, Garcia-Aznar JM. Unravelling cell migration: defining movement from the cell surface. Cell Adh Migr 2022; 16:25-64. [PMID: 35499121 PMCID: PMC9067518 DOI: 10.1080/19336918.2022.2055520] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Cell motility is essential for life and development. Unfortunately, cell migration is also linked to several pathological processes, such as cancer metastasis. Cells' ability to migrate relies on many actors. Cells change their migratory strategy based on their phenotype and the properties of the surrounding microenvironment. Cell migration is, therefore, an extremely complex phenomenon. Researchers have investigated cell motility for more than a century. Recent discoveries have uncovered some of the mysteries associated with the mechanisms involved in cell migration, such as intracellular signaling and cell mechanics. These findings involve different players, including transmembrane receptors, adhesive complexes, cytoskeletal components , the nucleus, and the extracellular matrix. This review aims to give a global overview of our current understanding of cell migration.
Collapse
Affiliation(s)
- Francisco Merino-Casallo
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Maria Jose Gomez-Benito
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Silvia Hervas-Raluy
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Jose Manuel Garcia-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
13
|
Jones CE, Sharick JT, Sizemore ST, Cukierman E, Strohecker AM, Leight JL. A miniaturized screening platform to identify novel regulators of extracellular matrix alignment. CANCER RESEARCH COMMUNICATIONS 2022; 2:1471-1486. [PMID: 36530465 PMCID: PMC9757767 DOI: 10.1158/2767-9764.crc-22-0157] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/03/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Extracellular matrix alignment contributes to metastasis in a number of cancers and is a known prognostic stromal factor; however, the mechanisms controlling matrix organization remain unclear. Cancer-associated fibroblasts (CAF) play a critical role in this process, particularly via matrix production and modulation of key signaling pathways controlling cell adhesion and contractility. Stroma normalization, as opposed to elimination, is a highly sought strategy, and screening for drugs that effectively alter extracellular matrix (ECM) alignment is a practical way to identify novel CAF-normalizing targets that modulate ECM organization. To meet this need, we developed a novel high-throughput screening platform in which fibroblast-derived matrices were produced in 384-well plates, imaged with automated confocal microscopy, and analyzed using a customized MATLAB script. This platform is a technical advance because it miniaturizes the assay, eliminates costly and time-consuming experimental steps, and streamlines data acquisition and analysis to enable high-throughput screening applications. As a proof of concept, this platform was used to screen a kinase inhibitor library to identify modulators of matrix alignment. A number of novel potential regulators were identified, including several receptor tyrosine kinases (c-MET, tropomyosin receptor kinase 1 (NTRK1), HER2/ERBB2) and the serine/threonine kinases protein kinase A, C, and G (PKA, PKC, and PKG). The expression of these regulators was analyzed in publicly available patient datasets to examine the association between stromal gene expression and patient outcomes.
Collapse
Affiliation(s)
- Caitlin E. Jones
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Joe T. Sharick
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
| | - Steven T. Sizemore
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Edna Cukierman
- Cancer Signaling and Epigenetics, The Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, Pennsylvania
| | - Anne Marie Strohecker
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Jennifer L. Leight
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
14
|
Ahmed A, Joshi IM, Goulet MR, Vidas JA, Byerley AM, Mansouri M, Day SW, Abhyankar VV. Microengineering 3D Collagen Hydrogels with Long-Range Fiber Alignment. J Vis Exp 2022:10.3791/64457. [PMID: 36156068 PMCID: PMC10203374 DOI: 10.3791/64457] [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] [Indexed: 10/31/2022] Open
Abstract
Aligned collagen I (COL1) fibers guide tumor cell motility, influence endothelial cell morphology, control stem cell differentiation, and are a hallmark of cardiac and musculoskeletal tissues. To study cell response to aligned microenvironments in vitro, several protocols have been developed to generate COL1 matrices with defined fiber alignment, including magnetic, mechanical, cell-based, and microfluidic methods. Of these, microfluidic approaches offer advanced capabilities such as accurate control over fluid flows and the cellular microenvironment. However, the microfluidic approaches to generate aligned COL1 matrices for advanced in vitro culture platforms have been limited to thin "mats" (<40 µm in thickness) of COL1 fibers that extend over distances less than 500 µm and are not conducive to 3D cell culture applications. Here, we present a protocol to fabricate 3D COL1 matrices (130-250 µm in thickness) with millimeter-scale regions of defined fiber alignment in a microfluidic device. This platform provides advanced cell culture capabilities to model structured tissue microenvironments by providing direct access to the micro-engineered matrix for cell culture.
Collapse
Affiliation(s)
- Adeel Ahmed
- Department of Biomedical Engineering, Kate Gleason College of Engineering, Rochester Institute of Technology
| | - Indranil M Joshi
- Department of Biomedical Engineering, Kate Gleason College of Engineering, Rochester Institute of Technology
| | - Madeleine R Goulet
- Department of Biomedical Engineering, Kate Gleason College of Engineering, Rochester Institute of Technology
| | - Justin A Vidas
- Department of Biomedical Engineering, Kate Gleason College of Engineering, Rochester Institute of Technology
| | - Ann M Byerley
- Department of Biomedical Engineering, Kate Gleason College of Engineering, Rochester Institute of Technology
| | - Mehran Mansouri
- Department of Biomedical Engineering, Kate Gleason College of Engineering, Rochester Institute of Technology
| | - Steven W Day
- Department of Biomedical Engineering, Kate Gleason College of Engineering, Rochester Institute of Technology
| | - Vinay V Abhyankar
- Department of Biomedical Engineering, Kate Gleason College of Engineering, Rochester Institute of Technology;
| |
Collapse
|
15
|
Ahmed A, Mansouri M, Joshi IM, Byerley AM, Day SW, Gaborski TR, Abhyankar VV. Local extensional flows promote long-range fiber alignment in 3D collagen hydrogels. Biofabrication 2022; 14. [PMID: 35735228 DOI: 10.1088/1758-5090/ac7824] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/13/2022] [Indexed: 02/07/2023]
Abstract
Randomly oriented type I collagen (COL1) fibers in the extracellular matrix are reorganized by biophysical forces into aligned domains extending several millimeters and with varying degrees of fiber alignment. These aligned fibers can transmit traction forces, guide tumor cell migration, facilitate angiogenesis, and influence tissue morphogenesis. To create aligned COL1 domains in microfluidic cell culture models, shear flows have been used to align thin COL1 matrices (<50µm in height) in a microchannel. However, there has been limited investigation into the role of shear flows in aligning 3D hydrogels (>130µm). Here, we show that pure shear flows do not induce fiber alignment in 3D atelo COL1 hydrogels, but the simple addition of local extensional flow promotes alignment that is maintained across several millimeters, with a degree of alignment directly related to the extensional strain rate. We further advance experimental capabilities by addressing the practical challenge of accessing a 3D hydrogel formed within a microchannel by introducing a magnetically coupled modular platform that can be released to expose the microengineered hydrogel. We demonstrate the platform's capability to pattern cells and fabricate multi-layered COL1 matrices using layer-by-layer fabrication and specialized modules. Our approach provides an easy-to-use fabrication method to achieve advanced hydrogel microengineering capabilities that combine fiber alignment with biofabrication capabilities.
Collapse
Affiliation(s)
- Adeel Ahmed
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, United States of America
| | - Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, United States of America
| | - Indranil M Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, United States of America
| | - Ann M Byerley
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, United States of America
| | - Steven W Day
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, United States of America
| | - Thomas R Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, United States of America
| | - Vinay V Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, United States of America
| |
Collapse
|
16
|
Burkel BM, Inman DR, Virumbrales-Muñoz M, Hoffmann EJ, Ponik SM. A Label-Free Segmentation Approach for Intravital Imaging of Mammary Tumor Microenvironment. JOURNAL OF VISUALIZED EXPERIMENTS : JOVE 2022:10.3791/63413. [PMID: 35695521 PMCID: PMC9327791 DOI: 10.3791/63413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The ability to visualize complex and dynamic physiological interactions between numerous cell types and the extracellular matrix (ECM) within a live tumor microenvironment is an important step toward understanding mechanisms that regulate tumor progression. While this can be accomplished through current intravital imaging techniques, it remains challenging due to the heterogeneous nature of tissues and the need for spatial context within the experimental observation. To this end, we have developed an intravital imaging workflow that pairs collagen second harmonic generation imaging, endogenous fluorescence from the metabolic co-factor NAD(P)H, and fluorescence lifetime imaging microscopy (FLIM) as a means to non-invasively compartmentalize the tumor microenvironment into basic domains of the tumor nest, the surrounding stroma or ECM, and the vasculature. This non-invasive protocol details the step-by-step process ranging from the acquisition of time-lapse images of mammary tumor models to post-processing analysis and image segmentation. The primary advantage of this workflow is that it exploits metabolic signatures to contextualize the dynamically changing live tumor microenvironment without the use of exogenous fluorescent labels, making it advantageous for human patient-derived xenograft (PDX) models and future clinical use where extrinsic fluorophores are not readily applicable.
Collapse
Affiliation(s)
- Brian M. Burkel
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison
| | - David R. Inman
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison
| | - María Virumbrales-Muñoz
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison,Department of Pathology, University of Wisconsin-Madison
| | - Erica J. Hoffmann
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison
| | - Suzanne M. Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison,Carbone Cancer Center, University of Wisconsin-Madison
| |
Collapse
|
17
|
Zanotelli MR, Zhang J, Ortiz I, Wang W, Chada NC, Reinhart-King CA. Highly motile cells are metabolically responsive to collagen density. Proc Natl Acad Sci U S A 2022; 119:e2114672119. [PMID: 35471912 PMCID: PMC9170068 DOI: 10.1073/pnas.2114672119] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 03/09/2022] [Indexed: 01/04/2023] Open
Abstract
Altered tissue mechanics and metabolism have gained significant attention as drivers of tumorigenesis, and mechanoresponsive metabolism has been implicated in migration and metastasis. However, heterogeneity in cell populations makes it difficult to link changes in behavior with metabolism, as individual cell behaviors are not necessarily reflected in population-based measurements. As such, the impact of increased collagen deposition, a tumor-associated collagen signature, on metabolism remains ambiguous. Here, we utilize a wide range of collagen densities to alter migration ability and study the bioenergetics of individual cells over time. Sorting cells based on their level of motility revealed energetics are a function of collagen density only for highly motile cells, not the entire population or cells with low motility. Changes in migration with increasing collagen density were correlated with cellular energetics, where matrix conditions most permissive to migration required less energy usage during movement and migrated more efficiently. These findings reveal a link between matrix mechanics, migratory phenotype, and bioenergetics and suggest that energetic costs are determined by the extracellular matrix and influence cell motility.
Collapse
Affiliation(s)
- Matthew R. Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Jian Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Ismael Ortiz
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | - Neil C. Chada
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235
| | | |
Collapse
|
18
|
Geiger F, Schnitzler LG, Brugger MS, Westerhausen C, Engelke H. Directed invasion of cancer cell spheroids inside 3D collagen matrices oriented by microfluidic flow in experiment and simulation. PLoS One 2022; 17:e0264571. [PMID: 35231060 PMCID: PMC8887745 DOI: 10.1371/journal.pone.0264571] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/14/2022] [Indexed: 01/07/2023] Open
Abstract
Invasion is strongly influenced by the mechanical properties of the extracellular matrix. Here, we use microfluidics to align fibers of a collagen matrix and study the influence of fiber orientation on invasion from a cancer cell spheroid. The microfluidic setup allows for highly oriented collagen fibers of tangential and radial orientation with respect to the spheroid, which can be described by finite element simulations. In invasion experiments, we observe a strong bias of invasion towards radial as compared to tangential fiber orientation. Simulations of the invasive behavior with a Brownian diffusion model suggest complete blockage of migration perpendicularly to fibers allowing for migration exclusively along fibers. This slows invasion toward areas with tangentially oriented fibers down, but does not prevent it.
Collapse
Affiliation(s)
- Florian Geiger
- Department of Chemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lukas G. Schnitzler
- Experimental Physics I, Institute of Physics, University of Augsburg, Augsburg, Germany
| | - Manuel S. Brugger
- Experimental Physics I, Institute of Physics, University of Augsburg, Augsburg, Germany
- Stiftung der Deutschen Wirtschaft (sdw) gGmbH, Berlin, Germany
| | - Christoph Westerhausen
- Experimental Physics I, Institute of Physics, University of Augsburg, Augsburg, Germany
- Physiology, Institute of Theoretical Medicine, University of Augsburg, Augsburg, Germany
- Center for NanoScience (CeNS), Munich, Germany
- * E-mail: (CW); (HE)
| | - Hanna Engelke
- Department of Chemistry, Ludwig-Maximilians-Universität München, Munich, Germany
- Center for NanoScience (CeNS), Munich, Germany
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Chemistry, University of Graz, Graz, Austria
- * E-mail: (CW); (HE)
| |
Collapse
|
19
|
Pamonag M, Hinson A, Burton EJ, Jafari N, Sales D, Babcock S, Basha R, Hu X, Kubow KE. Individual cells generate their own self-reinforcing contact guidance cues through local matrix fiber remodeling. PLoS One 2022; 17:e0265403. [PMID: 35333902 PMCID: PMC8956187 DOI: 10.1371/journal.pone.0265403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 03/01/2022] [Indexed: 11/18/2022] Open
Abstract
Directed cell migration arises from cells following a microenvironmental gradient (e.g. of a chemokine) or polarizing feature (e.g. a linear structure). However cells not only follow, but in many cases, also generate directionality cues by modifying their microenvironment. This bi-directional relationship is seen in the alignment of extracellular matrix (ECM) fibers ahead of invading cell masses. The forces generated by many migrating cells cause fiber alignment, which in turn promotes further migration in the direction of fiber alignment via contact guidance and durotaxis. While this positive-feedback relationship has been widely described for cells invading en masse, single cells are also able to align ECM fibers, as well as respond to contact guidance and durotaxis cues, and should therefore exhibit the same relationship. In this study, we directly tested this hypothesis by studying the migration persistence of individual HT-1080 fibrosarcoma cells migrating in photocrosslinked collagen matrices with limited remodeling potential. Our results demonstrate that this positive-feedback relationship is indeed a fundamental aspect of cell migration in fibrillar environments. We observed that the cells’ inability to align and condense fibers resulted in a decrease in persistence relative to cells in native collagen matrices and even relative to isotropic (glass) substrates. Further experiments involving 2D collagen and electrospun polymer scaffolds suggest that substrates composed of rigid, randomly oriented fibers reduce cells’ ability to follow another directionality cue by forcing them to meander to follow the available adhesive area (i.e. fibers). Finally, our results demonstrate that the bi-directional relationship between cell remodeling and migration is not a “dimensionality” effect, but a fundamental effect of fibrous substrate structure.
Collapse
Affiliation(s)
- Michael Pamonag
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Abigail Hinson
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Elisha J. Burton
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Nojan Jafari
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Dominic Sales
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Sarah Babcock
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Rozlan Basha
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
| | - Xiaofeng Hu
- Department of Chemistry & Biochemistry and Center for Materials Science, James Madison University, Harrisonburg, Virginia, United States of America
| | - Kristopher E. Kubow
- Department of Biology, James Madison University, Harrisonburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
20
|
Nguyen RY, Xiao H, Gong X, Arroyo A, Cabral AT, Fischer TT, Flores KM, Zhang X, Robert ME, Ehrlich BE, Mak M. Cytoskeletal dynamics regulates stromal invasion behavior of distinct liver cancer subtypes. Commun Biol 2022; 5:202. [PMID: 35241781 PMCID: PMC8894393 DOI: 10.1038/s42003-022-03121-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/08/2022] [Indexed: 02/07/2023] Open
Abstract
Drug treatment against liver cancer has limited efficacy due to heterogeneous response among liver cancer subtypes. In addition, the functional biophysical phenotypes which arise from this heterogeneity and contribute to aggressive invasive behavior remain poorly understood. This study interrogated how heterogeneity in liver cancer subtypes contributes to differences in invasive phenotypes and drug response. Utilizing histological analysis, quantitative 2D invasion metrics, reconstituted 3D hydrogels, and bioinformatics, our study linked cytoskeletal dynamics to differential invasion profiles and drug resistance in liver cancer subtypes. We investigated cytoskeletal regulation in 2D and 3D culture environments using two liver cancer cell lines, SNU-475 and HepG2, chosen for their distinct cytoskeletal features and invasion profiles. For SNU-475 cells, a model for aggressive liver cancer, many cytoskeletal inhibitors abrogated 2D migration but only some suppressed 3D migration. For HepG2 cells, cytoskeletal inhibition did not significantly affect 3D migration but did affect proliferative capabilities and spheroid core growth. This study highlights cytoskeleton driven phenotypic variation, their consequences and coexistence within the same tumor, as well as efficacy of targeting biophysical phenotypes that may be masked in traditional screens against tumor growth.
Collapse
Affiliation(s)
- Ryan Y Nguyen
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Hugh Xiao
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xiangyu Gong
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Alfredo Arroyo
- Department of Pharmacology, Yale University, New Haven, CT, USA
| | - Aidan T Cabral
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Tom T Fischer
- Department of Pharmacology, Yale University, New Haven, CT, USA
- Institute of Pharmacology, University of Heidelberg, Heidelberg, Germany
| | - Kaitlin M Flores
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Marie E Robert
- Department of Pathology, Yale University, New Haven, CT, USA
| | | | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
21
|
Ray A, Callaway MK, Rodríguez-Merced NJ, Crampton AL, Carlson M, Emme KB, Ensminger EA, Kinne AA, Schrope JH, Rasmussen HR, Jiang H, DeNardo DG, Wood DK, Provenzano PP. Stromal architecture directs early dissemination in pancreatic ductal adenocarcinoma. JCI Insight 2021; 7:150330. [PMID: 34914633 PMCID: PMC8855836 DOI: 10.1172/jci.insight.150330] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 12/10/2021] [Indexed: 12/02/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is an extremely metastatic and lethal disease. Here, in both murine and human PDA, we demonstrate that extracellular matrix architecture regulates cell extrusion and subsequent invasion from intact ductal structures through tumor-associated collagen signatures (TACS). This results in early dissemination from histologically premalignant lesions and continual invasion from well-differentiated disease, and it suggests TACS as a biomarker to aid in the pathologic assessment of early disease. Furthermore, we show that pancreatitis results in invasion-conducive architectures, thus priming the stroma prior to malignant disease. Analysis in potentially novel microfluidic-derived microtissues and in vivo demonstrates decreased extrusion and invasion following focal adhesion kinase (FAK) inhibition, consistent with decreased metastasis. Thus, data suggest that targeting FAK or strategies to reengineer and normalize tumor microenvironments may have roles not only in very early disease, but also for limiting continued dissemination from unresectable disease. Likewise, it may be beneficial to employ stroma-targeting strategies to resolve precursor diseases such as pancreatitis in order to remove stromal architectures that increase risk for early dissemination.
Collapse
Affiliation(s)
- Arja Ray
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Mackenzie K Callaway
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Nelson J Rodríguez-Merced
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Alexandra L Crampton
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Marjorie Carlson
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Kenneth B Emme
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Ethan A Ensminger
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Alexander A Kinne
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Jonathan H Schrope
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Haley R Rasmussen
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Hong Jiang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States of America
| | - David G DeNardo
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, United States of America
| | - David K Wood
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| | - Paolo P Provenzano
- Department of Biomedical Engineeirng, University of Minnesota, Minneapolis, United States of America
| |
Collapse
|
22
|
Ray A, Provenzano PP. Aligned forces: Origins and mechanisms of cancer dissemination guided by extracellular matrix architecture. Curr Opin Cell Biol 2021; 72:63-71. [PMID: 34186415 PMCID: PMC8530881 DOI: 10.1016/j.ceb.2021.05.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/14/2022]
Abstract
Organized extracellular matrix (ECM), in the form of aligned architectures, is a critical mediator of directed cancer cell migration by contact guidance, leading to metastasis in solid tumors. Current models suggest anisotropic force generation through the engagement of key adhesion and cytoskeletal complexes drives contact-guided migration. Likewise, disrupting the balance between cell-cell and cell-ECM forces, driven by ECM engagement for cells at the tumor-stromal interface, initiates and drives local invasion. Furthermore, processes such as traction forces exerted by cancer and stromal cells, spontaneous reorientation of matrix-producing fibroblasts, and direct binding of ECM modifying proteins lead to the emergence of collagen alignment in tumors. Thus, as we obtain a deeper understanding of the origins of ECM alignment and the mechanisms by which it is maintained to direct invasion, we are poised to use the new paradigm of stroma-targeted therapies to disrupt this vital axis of disease progression in solid tumors.
Collapse
Affiliation(s)
- Arja Ray
- Department of Pathology, University of California, San Francisco, USA.
| | - Paolo P Provenzano
- Department of Biomedical Engineering, University of Minnesota, USA; University of Minnesota Physical Sciences in Oncology Center, USA; Masonic Cancer Center, University of Minnesota, USA; Institute for Engineering in Medicine, University of Minnesota, USA; Stem Cell Institute, University of Minnesota, USA.
| |
Collapse
|
23
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
24
|
Directional cues in the tumor microenvironment due to cell contraction against aligned collagen fibers. Acta Biomater 2021; 129:96-109. [PMID: 33965625 PMCID: PMC8848478 DOI: 10.1016/j.actbio.2021.04.053] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023]
Abstract
It is well established that collagen alignment in the breast tumor microenvironment provides biophysical cues to drive disease progression. Numerous mechanistic studies have demonstrated that tumor cell behavior is driven by the architecture and stiffness of the collagen matrix. However, the mechanical properties within a 3D collagen microenvironment, particularly at the scale of the cell, remain poorly defined. To investigate cell-scale mechanical cues with respect to local collagen architecture, we employed a combination of intravital imaging of the mammary tumor microenvironment and a 3D collagen gel system with both acellular pNIPAAm microspheres and MDA-MB-231 breast carcinoma cells. Within the in vivo tumor microenvironment, the displacement of collagen fiber was identified in response to tumor cells migrating through the stromal matrix. To further investigate cell-scale stiffness in aligned fiber architectures and the propagation of cell-induced fiber deformations, precise control of collagen architecture was coupled with innovative methodology to measure mechanical properties of the collagen fiber network. This method revealed up to a 35-fold difference in directional cell-scale stiffness resulting from contraction against aligned fibers. Furthermore, the local anisotropy of the matrix dramatically altered the rate at which contractility-induced fiber displacements decayed over distance. Together, our results reveal mechanical properties in aligned matrices that provide dramatically different cues to the cell in perpendicular directions. These findings are supported by the mechanosensing behavior of tumor cells and have important implications for cell-cell communication within the tissue microenvironment. STATEMENT OF SIGNIFICANCE: It is widely appreciated that the architecture of the extracellular matrix impacts cellular behavior in normal and disease states. Numerous studies have determined the fundamental role of collagen matrix architecture on cellular mechanosensing, but effectively quantifying anisotropic mechanical properties of the collagen matrix at the cell-scale remains challenging. Here, we developed innovative methodology to discover that collagen alignment results in a 35-fold difference in cell-scale stiffness and alters contractile force transmission through the fiber network. Furthermore, we identified bias in cell response along the axis of alignment, where local stiffness is highest. Overall, our results define cell-scale stiffness and fiber deformations due to collagen architecture that may instruct cell communication within a broad range of tissue microenvironments.
Collapse
|
25
|
Cell contact guidance via sensing anisotropy of network mechanical resistance. Proc Natl Acad Sci U S A 2021; 118:2024942118. [PMID: 34266950 DOI: 10.1073/pnas.2024942118] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Despite the ubiquitous importance of cell contact guidance, the signal-inducing contact guidance of mammalian cells in an aligned fibril network has defied elucidation. This is due to multiple interdependent signals that an aligned fibril network presents to cells, including, at least, anisotropy of adhesion, porosity, and mechanical resistance. By forming aligned fibrin gels with the same alignment strength, but cross-linked to different extents, the anisotropic mechanical resistance hypothesis of contact guidance was tested for human dermal fibroblasts. The cross-linking was shown to increase the mechanical resistance anisotropy, without detectable change in network microstructure and without change in cell adhesion to the cross-linked fibrin gel. This methodology thus isolated anisotropic mechanical resistance as a variable for fixed anisotropy of adhesion and porosity. The mechanical resistance anisotropy |Y*| -1 - |X*| -1 increased over fourfold in terms of the Fourier magnitudes of microbead displacement |X*| and |Y*| at the drive frequency with respect to alignment direction Y obtained by optical forces in active microrheology. Cells were found to exhibit stronger contact guidance in the cross-linked gels possessing greater mechanical resistance anisotropy: the cell anisotropy index based on the tensor of cell orientation, which has a range 0 to 1, increased by 18% with the fourfold increase in mechanical resistance anisotropy. We also show that modulation of adhesion via function-blocking antibodies can modulate the guidance response, suggesting a concomitant role of cell adhesion. These results indicate that fibroblasts can exhibit contact guidance in aligned fibril networks by sensing anisotropy of network mechanical resistance.
Collapse
|
26
|
Hiraki HL, Matera DL, Rose MJ, Kent RN, Todd CW, Stout ME, Wank AE, Schiavone MC, DePalma SJ, Zarouk AA, Baker BM. Magnetic Alignment of Electrospun Fiber Segments Within a Hydrogel Composite Guides Cell Spreading and Migration Phenotype Switching. Front Bioeng Biotechnol 2021; 9:679165. [PMID: 34222216 PMCID: PMC8242362 DOI: 10.3389/fbioe.2021.679165] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/10/2021] [Indexed: 01/09/2023] Open
Abstract
Fibrous extracellular matrix (ECM) proteins provide mechanical structure and adhesive scaffolding to resident cells within stromal tissues. Aligned ECM fibers play an important role in directing morphogenetic processes, supporting mechanical loads, and facilitating cell migration. Various methods have been developed to align matrix fibers in purified biopolymer hydrogels, such as type I collagen, including flow-induced alignment, uniaxial tensile deformation, and magnetic particles. However, purified biopolymers have limited orthogonal tunability of biophysical cues including stiffness, fiber density, and fiber alignment. Here, we generate synthetic, cell-adhesive fiber segments of the same length-scale as stromal fibrous proteins through electrospinning. Superparamagnetic iron oxide nanoparticles (SPIONs) embedded in synthetic fiber segments enable magnetic field induced alignment of fibers within an amorphous bulk hydrogel. We find that SPION density and magnetic field strength jointly influence fiber alignment and identify conditions to control the degree of alignment. Tuning fiber length allowed the alignment of dense fibrous hydrogel composites without fiber entanglement or regional variation in the degree of alignment. Functionalization of fiber segments with cell adhesive peptides induced tendon fibroblasts to adopt a uniaxial morphology akin to within native tendon. Furthermore, we demonstrate the utility of this hydrogel composite to direct multicellular migration from MCF10A spheroids and find that fiber alignment prompts invading multicellular strands to separate into disconnected single cells and multicellular clusters. These magnetic fiber segments can be readily incorporated into other natural and synthetic hydrogels and aligned with inexpensive and easily accessible rare earth magnets, without the need for specialized equipment. 3D hydrogel composites where stiffness/crosslinking, fiber density, and fiber alignment can be orthogonally tuned may provide insights into morphogenetic and pathogenic processes that involve matrix fiber alignment and can enable systematic investigation of the individual contribution of each biophysical cue to cell behavior.
Collapse
Affiliation(s)
- Harrison L. Hiraki
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Daniel L. Matera
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Michael J. Rose
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Robert N. Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Connor W. Todd
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Mark E. Stout
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Anya E. Wank
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Maria C. Schiavone
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Alexander A. Zarouk
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
27
|
Abdollahiyan P, Oroojalian F, Baradaran B, de la Guardia M, Mokhtarzadeh A. Advanced mechanotherapy: Biotensegrity for governing metastatic tumor cell fate via modulating the extracellular matrix. J Control Release 2021; 335:596-618. [PMID: 34097925 DOI: 10.1016/j.jconrel.2021.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022]
Abstract
Mechano-transduction is the procedure of mechanical stimulus translation via cells, among substrate shear flow, topography, and stiffness into a biochemical answer. TAZ and YAP are transcriptional coactivators which are recognized as relay proteins that promote mechano-transduction within the Hippo pathway. With regard to healthy cells in homeostasis, mechano-transduction regularly restricts proliferation, and TAZ and YAP are totally inactive. During cancer development a YAP/TAZ - stimulating positive response loop is formed between the growing tumor and the stiffening ECM. As tumor developments, local stromal and cancerous cells take advantage of mechanotransduction to enhance proliferation, induce their migratory into remote tissues, and promote chemotherapeutic resistance. As a newly progresses paradigm, nanoparticle-conjunctions (such as magnetic nanoparticles, and graphene derivatives nanoparticles) hold significant promises for remote regulation of cells and their relevant events at molecular scale. Despite outstanding developments in employing nanoparticles for drug targeting studies, the role of nanoparticles on cellular behaviors (proliferation, migration, and differentiation) has still required more evaluations in the field of mechanotherapy. In this paper, the in-depth contribution of mechano-transduction is discussed during tumor progression, and how these consequences can be evaluated in vitro.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Valencia, Spain
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
28
|
Ahmed A, Joshi IM, Mansouri M, Ahamed NNN, Hsu MC, Gaborski TR, Abhyankar VV. Engineering fiber anisotropy within natural collagen hydrogels. Am J Physiol Cell Physiol 2021; 320:C1112-C1124. [PMID: 33852366 PMCID: PMC8285641 DOI: 10.1152/ajpcell.00036.2021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/14/2022]
Abstract
It is well known that biophysical properties of the extracellular matrix (ECM), including stiffness, porosity, composition, and fiber alignment (anisotropy), play a crucial role in controlling cell behavior in vivo. Type I collagen (collagen I) is a ubiquitous structural component in the ECM and has become a popular hydrogel material that can be tuned to replicate the mechanical properties found in vivo. In this review article, we describe popular methods to create 2-D and 3-D collagen I hydrogels with anisotropic fiber architectures. We focus on methods that can be readily translated from engineering and materials science laboratories to the life-science community with the overall goal of helping to increase the physiological relevance of cell culture assays.
Collapse
Affiliation(s)
- Adeel Ahmed
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
| | - Indranil M Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, New York
| | - Mehran Mansouri
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
| | - Nuzhet N N Ahamed
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
| | - Meng-Chun Hsu
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
| | - Thomas R Gaborski
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, New York
| | - Vinay V Abhyankar
- Department of Microsystems Engineering, Rochester Institute of Technology, Rochester, New York
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, New York
| |
Collapse
|
29
|
Hapach LA, Carey SP, Schwager SC, Taufalele PV, Wang W, Mosier JA, Ortiz-Otero N, McArdle TJ, Goldblatt ZE, Lampi MC, Bordeleau F, Marshall JR, Richardson IM, Li J, King MR, Reinhart-King CA. Phenotypic Heterogeneity and Metastasis of Breast Cancer Cells. Cancer Res 2021; 81:3649-3663. [PMID: 33975882 DOI: 10.1158/0008-5472.can-20-1799] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 02/02/2021] [Accepted: 05/05/2021] [Indexed: 11/16/2022]
Abstract
Although intratumoral genomic heterogeneity can impede cancer research and treatment, less is known about the effects of phenotypic heterogeneities. To investigate the role of cell migration heterogeneities in metastasis, we phenotypically sorted metastatic breast cancer cells into two subpopulations based on migration ability. Although migration is typically considered to be associated with metastasis, when injected orthotopically in vivo, the weakly migratory subpopulation metastasized significantly more than the highly migratory subpopulation. To investigate the mechanism behind this observation, both subpopulations were assessed at each stage of the metastatic cascade, including dissemination from the primary tumor, survival in the circulation, extravasation, and colonization. Although both subpopulations performed each step successfully, weakly migratory cells presented as circulating tumor cell (CTC) clusters in the circulation, suggesting clustering as one potential mechanism behind the increased metastasis of weakly migratory cells. RNA sequencing revealed weakly migratory subpopulations to be more epithelial and highly migratory subpopulations to be more mesenchymal. Depletion of E-cadherin expression from weakly migratory cells abrogated metastasis. Conversely, induction of E-cadherin expression in highly migratory cells increased metastasis. Clinical patient data and blood samples showed that CTC clustering and E-cadherin expression are both associated with worsened patient outcome. This study demonstrates that deconvolving phenotypic heterogeneities can reveal fundamental insights into metastatic progression. More specifically, these results indicate that migratory ability does not necessarily correlate with metastatic potential and that E-cadherin promotes metastasis in phenotypically sorted breast cancer cell subpopulations by enabling CTC clustering. SIGNIFICANCE: This study employs phenotypic cell sorting for migration to reveal a weakly migratory, highly metastatic breast cancer cell subpopulation regulated by E-cadherin, highlighting the dichotomy between cancer cell migration and metastasis.
Collapse
Affiliation(s)
- Lauren A Hapach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.,Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Shawn P Carey
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Samantha C Schwager
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Paul V Taufalele
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Wenjun Wang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Nerymar Ortiz-Otero
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | | | - Zachary E Goldblatt
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Marsha C Lampi
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Francois Bordeleau
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.,CHU de Québec-Université Laval Research Center, Université Laval Cancer Research Center, Québec, Canada
| | - Jocelyn R Marshall
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Isaac M Richardson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Jiahe Li
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
30
|
Wu Y, Zanotelli MR, Zhang J, Reinhart-King CA. Matrix-driven changes in metabolism support cytoskeletal activity to promote cell migration. Biophys J 2021; 120:1705-1717. [PMID: 33705759 PMCID: PMC8204337 DOI: 10.1016/j.bpj.2021.02.044] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 01/21/2023] Open
Abstract
The microenvironment provides both active and passive mechanical cues that regulate cell morphology, adhesion, migration, and metabolism. Although the cellular response to those mechanical cues often requires energy-intensive actin cytoskeletal remodeling and actomyosin contractility, it remains unclear how cells dynamically adapt their metabolic activity to altered mechanical cues to support migration. Here, we investigated the changes in cellular metabolic activity in response to different two-dimensional and three-dimensional microenvironmental conditions and how these changes relate to cytoskeletal activity and migration. Utilizing collagen micropatterning on polyacrylamide gels, intracellular energy levels and oxidative phosphorylation were found to be correlated with cell elongation and spreading and necessary for membrane ruffling. To determine whether this relationship holds in more physiological three-dimensional matrices, collagen matrices were used to show that intracellular energy state was also correlated with protrusive activity and increased with matrix density. Pharmacological inhibition of oxidative phosphorylation revealed that cancer cells rely on oxidative phosphorylation to meet the elevated energy requirements for protrusive activity and migration in denser matrices. Together, these findings suggest that mechanical regulation of cytoskeletal activity during spreading and migration by the physical microenvironment is driven by an altered metabolic profile.
Collapse
Affiliation(s)
- Yusheng Wu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Matthew R Zanotelli
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jian Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
31
|
Abstract
The extracellular matrix is a fundamental, core component of all tissues and organs, and is essential for the existence of multicellular organisms. From the earliest stages of organism development until death, it regulates and fine-tunes every cellular process in the body. In cancer, the extracellular matrix is altered at the biochemical, biomechanical, architectural and topographical levels, and recent years have seen an exponential increase in the study and recognition of the importance of the matrix in solid tumours. Coupled with the advancement of new technologies to study various elements of the matrix and cell-matrix interactions, we are also beginning to see the deployment of matrix-centric, stromal targeting cancer therapies. This Review touches on many of the facets of matrix biology in solid cancers, including breast, pancreatic and lung cancer, with the aim of highlighting some of the emerging interactions of the matrix and influences that the matrix has on tumour onset, progression and metastatic dissemination, before summarizing the ongoing work in the field aimed at developing therapies to co-target the matrix in cancer and cancer metastasis.
Collapse
Affiliation(s)
- Thomas R Cox
- The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
32
|
Tang H, Yi B, Wang X, Shen Y, Zhang Y. Understanding the cellular responses based on low-density electrospun fiber networks. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111470. [PMID: 33321594 DOI: 10.1016/j.msec.2020.111470] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/14/2020] [Accepted: 08/25/2020] [Indexed: 01/12/2023]
Abstract
Fibers produced from electrospinning are well-known to be extremely fine with diameters ranging from tens of nanometers to a few microns. Such ultrafine fibers not only allow for engineering scaffolds resembling the ultrastructure of the native extracellular matrix, but also offer possibility to explore the remodeling behavior of cells in vitro, due to their mechanically 'adequate' softness endowed by their ultrafine fineness. However, the remodeling effect of cells on the biomimicking fibrous substrates remains to be understood, because the crisscrossing and entangling among nanofibers in those tightly packed fibrous mats ultimately lead to merely a topological phenomenon, similar to that of the nanofiber-like topography embossed on the surface of a solid matter. In this study, the effect of nanofiber density on cellular response behavior was investigated by reducing the density of electrospun fiber networks. Using polycaprolactone (PCL) as a model polymer, randomly oriented fiber networks with various densities, namely, 37.7 ± 16.3 μg/cm2 (D1), 103.8 ± 16.3 μg/cm2 (D2), 198.2 ± 40.0 μg/cm2 (D3), and 471.8 ± 32.7 μg/cm2 (D4), were prepared by electrospinning for varied collection durations (10 s, 50 s, 100 s, and 10 min, respectively). By examining the responsive behavior of the human induced pluripotent stem cell-derived mesenchymal stem cells (hiPS-MSCs) cultured on these nanofibrous networks, we showed that the fiber network with a moderate density (D2) is beneficial to the cell attachment, spreading, actin polymerization, contractility and migration. There also showed an increased tendency in nuclear localization of the Yes-associated protein (YAP) and subsequent activation of YAP responsive gene transcription, and cell proliferation and collagen synthesis were also enhanced on the D2. However, further increasing the fiber density (D3, D4) gave rise to weakened induction effect of fibers on the cellular responses. These results enrich our understanding on the effect of fiber density on cell behavior, and disclose the dependence of cellular responses on fiber density. This study paves the way to precisely design biomimetic fibrous scaffolds for achieving enhanced cell-scaffold interactions and tissue regeneration.
Collapse
Affiliation(s)
- Han Tang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Bingcheng Yi
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Xianliu Wang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Yanbing Shen
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China
| | - Yanzhong Zhang
- College of Chemistry, Chemical Engineering & Biotechnology, Donghua University, Shanghai 201620, China; Key Lab of Science & Technology of Eco-Textile, Ministry of Education, Donghua University, Shanghai 201620, China; Shanghai Key Laboratory of Tissue Engineering, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou 310058, China.
| |
Collapse
|
33
|
Damaghi M, Mori H, Byrne S, Xu L, Chen T, Johnson J, Gallant ND, Marusyk A, Borowsky AD, Gillies RJ. Collagen production and niche engineering: A novel strategy for cancer cells to survive acidosis in DCIS and evolve. Evol Appl 2020; 13:2689-2703. [PMID: 33294017 PMCID: PMC7691473 DOI: 10.1111/eva.13075] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/09/2020] [Accepted: 07/12/2020] [Indexed: 12/31/2022] Open
Abstract
Growing tumors are dynamic and nonlinear ecosystems, wherein cancer cells adapt to their local microenvironment, and these adaptations further modify the environment, inducing more changes. From nascent intraductal neoplasms to disseminated metastatic disease, several levels of evolutionary adaptations and selections occur. Here, we focus on one example of such an adaptation mechanism, namely, "niche construction" promoted by adaptation to acidosis, which is a metabolic adaptation to the early harsh environment in intraductal neoplasms. The avascular characteristics of ductal carcinoma in situ (DCIS) make the periluminal volume profoundly acidic, and cancer cells must adapt to this to survive. Based on discovery proteomics, we hypothesized that a component of acid adaptation involves production of collagen by pre-cancer cells that remodels the extracellular matrix (ECM) and stabilizes cells under acid stress. The proteomic data were surprising as collagen production and deposition are commonly believed to be the responsibility of mesenchymally derived fibroblasts, and not cells of epithelial origin. Subsequent experiments in 3D culture, spinning disk and second harmonic generation microscopy of DCIS lesions in patients' samples are concordant. Collagen production assay by acid-adapted cells in vitro demonstrated that the mechanism of induction involves the RAS and SMAD pathways. Secretome analyses show upregulation of ECM remodeling enzymes such as TGM2 and LOXL2 that are collagen crosslinkers. These data strongly indicate that acidosis in incipient cancers induces collagen production by cancer cells and support the hypothesis that this adaptation initiates a tumor-permissive microenvironment promoting survival and growth of nascent cancers.
Collapse
Affiliation(s)
- Mehdi Damaghi
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
- Department of Oncologic SciencesMorsani College of MedicineUniversity of South FloridaTampaFLUSA
| | - Hidetoshi Mori
- Center for Immunology and Infectious DiseasesComprehensive Cancer CenterDepartment of Pathology and Laboratory MedicineSchool of MedicineUniversity of California, DavisSacramentoCAUSA
| | - Samantha Byrne
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Liping Xu
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Tingan Chen
- Analytic Microscopy CoreMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Joseph Johnson
- Analytic Microscopy CoreMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Nathan D. Gallant
- Department of Mechanical EngineeringUniversity of South FloridaTampaFLUSA
| | - Andriy Marusyk
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| | - Alexander D. Borowsky
- Center for Immunology and Infectious DiseasesComprehensive Cancer CenterDepartment of Pathology and Laboratory MedicineSchool of MedicineUniversity of California, DavisSacramentoCAUSA
| | - Robert J. Gillies
- Department of Cancer PhysiologyMoffitt Cancer Center and Research InstituteTampaFLUSA
| |
Collapse
|
34
|
Micek HM, Visetsouk MR, Masters KS, Kreeger PK. Engineering the Extracellular Matrix to Model the Evolving Tumor Microenvironment. iScience 2020; 23:101742. [PMID: 33225247 PMCID: PMC7666341 DOI: 10.1016/j.isci.2020.101742] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Clinical evidence supports a role for the extracellular matrix (ECM) in cancer risk and prognosis across multiple tumor types, and numerous studies have demonstrated that individual ECM components impact key hallmarks of tumor progression (e.g., proliferation, migration, angiogenesis). However, the ECM is a complex network of fibrillar proteins, glycoproteins, and proteoglycans that undergoes dramatic changes in composition and organization during tumor development. In this review, we will highlight how engineering approaches can be used to examine the impact of changes in tissue architecture, ECM composition (i.e., identity and levels of individual ECM components), and cellular- and tissue-level mechanics on tumor progression. In addition, we will discuss recently developed methods to model the ECM that have not yet been applied to the study of cancer.
Collapse
Affiliation(s)
- Hannah M. Micek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Mike R. Visetsouk
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kristyn S. Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Pamela K. Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
35
|
Hayn A, Fischer T, Mierke CT. Inhomogeneities in 3D Collagen Matrices Impact Matrix Mechanics and Cancer Cell Migration. Front Cell Dev Biol 2020; 8:593879. [PMID: 33251219 PMCID: PMC7674772 DOI: 10.3389/fcell.2020.593879] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Cell motility under physiological and pathological conditions including malignant progression of cancer and subsequent metastasis are founded on environmental confinements. During the last two decades, three-dimensional cell migration has been studied mostly by utilizing biomimetic extracellular matrix models. In the majority of these studies, the in vitro collagen scaffolds are usually assumed to be homogenous, as they consist commonly of one specific type of collagen, such as collagen type I, isolated from one species. These collagen matrices should resemble in vivo extracellular matrix scaffolds physiologically, however, mechanical phenotype and functional reliability have been addressed poorly due to certain limitations based on the assumption of homogeneity. How local variations of extracellular matrix structure impact matrix mechanics and cell migration is largely unknown. Here, we hypothesize that local inhomogeneities alter cell movement due to alterations in matrix mechanics, as they frequently occur in in vivo tissue scaffolds and were even changed in diseased tissues. To analyze the effect of structural inhomogeneities on cell migration, we used a mixture of rat tail and bovine dermal collagen type I as well as pure rat and pure bovine collagens at four different concentrations to assess three-dimensional scaffold inhomogeneities. Collagen type I from rat self-assembled to elongated fibrils, whereas bovine collagen tended to build node-shaped inhomogeneous scaffolds. We have shown that the elastic modulus determined with atomic force microscopy in combination with pore size analysis using confocal laser scanning microscopy revealed distinct inhomogeneities within collagen matrices. We hypothesized that elastic modulus and pore size govern cancer cell invasion in three-dimensional collagen matrices. In fact, invasiveness of three breast cancer cell types is altered due to matrix-type and concentration indicating that these two factors are crucial for cellular invasiveness. Our findings revealed that local matrix scaffold inhomogeneity is another crucial parameter to explain differences in cell migration, which not solely depended on pore size and stiffness of the collagen matrices. With these three distinct biophysical parameters, characterizing structure and mechanics of the studied collagen matrices, we were able to explain differences in the invasion behavior of the studied cancer cell lines in dependence of the used collagen model.
Collapse
Affiliation(s)
- Alexander Hayn
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| | - Tony Fischer
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| | - Claudia Tanja Mierke
- Biological Physics Division, Faculty of Physics and Earth Sciences, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Leipzig, Germany
| |
Collapse
|
36
|
Lowen JM, Leach JK. Functionally graded biomaterials for use as model systems and replacement tissues. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1909089. [PMID: 33456431 PMCID: PMC7810245 DOI: 10.1002/adfm.201909089] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Indexed: 05/03/2023]
Abstract
The heterogeneity of native tissues requires complex materials to provide suitable substitutes for model systems and replacement tissues. Functionally graded materials have the potential to address this challenge by mimicking the gradients in heterogeneous tissues such as porosity, mineralization, and fiber alignment to influence strength, ductility, and cell signaling. Advancements in microfluidics, electrospinning, and 3D printing enable the creation of increasingly complex gradient materials that further our understanding of physiological gradients. The combination of these methods enables rapid prototyping of constructs with high spatial resolution. However, successful translation of these gradients requires both spatial and temporal presentation of cues to model the complexity of native tissues that few materials have demonstrated. This review highlights recent strategies to engineer functionally graded materials for the modeling and repair of heterogeneous tissues, together with a description of how cells interact with various gradients.
Collapse
Affiliation(s)
- Jeremy M. Lowen
- Department of Biomedical Engineering, University of California, Davis, CA, 95616
| | - J. Kent Leach
- Department of Biomedical Engineering, University of California, Davis, CA, 95616
- Department of Orthopaedic Surgery, UC Davis Health, Sacramento, CA 95817
| |
Collapse
|
37
|
Becsky D, Szabo K, Gyulai-Nagy S, Gajdos T, Bartos Z, Balind A, Dux L, Horvath P, Erdelyi M, Homolya L, Keller-Pinter A. Syndecan-4 Modulates Cell Polarity and Migration by Influencing Centrosome Positioning and Intracellular Calcium Distribution. Front Cell Dev Biol 2020; 8:575227. [PMID: 33178691 PMCID: PMC7593626 DOI: 10.3389/fcell.2020.575227] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022] Open
Abstract
Efficient cell migration requires cellular polarization, which is characterized by the formation of leading and trailing edges, appropriate positioning of the nucleus and reorientation of the Golgi apparatus and centrosomes toward the leading edge. Migration also requires the development of an asymmetrical front-to-rear calcium (Ca2+) gradient to regulate focal adhesion assembly and actomyosin contractility. Here we demonstrate that silencing of syndecan-4, a transmembrane heparan sulfate proteoglycan, interferes with the correct polarization of migrating mammalian myoblasts (i.e., activated satellite stem cells). In particular, syndecan-4 knockdown completely abolished the intracellular Ca2+ gradient, abrogated centrosome reorientation and thus decreased cell motility, demonstrating the role of syndecan-4 in cell polarity. Additionally, syndecan-4 exhibited a polarized distribution during migration. Syndecan-4 knockdown cells exhibited decreases in the total movement distance during directional migration, maximum and vectorial distances from the starting point, as well as average and maximum cell speeds. Super-resolution direct stochastic optical reconstruction microscopy images of syndecan-4 knockdown cells revealed nanoscale changes in the actin cytoskeletal architecture, such as decreases in the numbers of branches and individual branch lengths in the lamellipodia of the migrating cells. Given the crucial importance of myoblast migration during embryonic development and postnatal muscle regeneration, we conclude that our results could facilitate an understanding of these processes and the general role of syndecan-4 during cell migration.
Collapse
Affiliation(s)
- Daniel Becsky
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Kitti Szabo
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Szuzina Gyulai-Nagy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tamas Gajdos
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Zsuzsa Bartos
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, Budapest, Hungary
| | - Arpad Balind
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Laszlo Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Peter Horvath
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Miklos Erdelyi
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Laszlo Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, Budapest, Hungary
| | - Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
38
|
Chen YQ, Kuo JC, Wei MT, Wu MC, Yang MH, Chiou A. Fibroblast Promotes Head and Neck Squamous Cell Carcinoma Cell Invasion through Mechanical Barriers in 3D Collagen Microenvironments. ACS APPLIED BIO MATERIALS 2020; 3:6419-6429. [DOI: 10.1021/acsabm.0c00603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Yin-Quan Chen
- Cancer Progression Research Center, National Yang-Ming University, Taipei 11221, Taiwan
| | - Jean-Cheng Kuo
- Cancer Progression Research Center, National Yang-Ming University, Taipei 11221, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Ming-Tzo Wei
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Ming-Chung Wu
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 11221, Taiwan
| | - Muh-Hwa Yang
- Cancer Progression Research Center, National Yang-Ming University, Taipei 11221, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 11221, Taiwan
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Arthur Chiou
- Institute of Biophotonics, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
39
|
Maxian O, Mogilner A, Strychalski W. Computational estimates of mechanical constraints on cell migration through the extracellular matrix. PLoS Comput Biol 2020; 16:e1008160. [PMID: 32853248 PMCID: PMC7480866 DOI: 10.1371/journal.pcbi.1008160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 09/09/2020] [Accepted: 07/17/2020] [Indexed: 12/17/2022] Open
Abstract
Cell migration through a three-dimensional (3D) extracellular matrix (ECM) underlies important physiological phenomena and is based on a variety of mechanical strategies depending on the cell type and the properties of the ECM. By using computer simulations of the cell’s mid-plane, we investigate two such migration mechanisms—‘push-pull’ (forming a finger-like protrusion, adhering to an ECM node, and pulling the cell body forward) and ‘rear-squeezing’ (pushing the cell body through the ECM by contracting the cell cortex and ECM at the cell rear). We present a computational model that accounts for both elastic deformation and forces of the ECM, an active cell cortex and nucleus, and for hydrodynamic forces and flow of the extracellular fluid, cytoplasm, and nucleoplasm. We find that relations between three mechanical parameters—the cortex’s contractile force, nuclear elasticity, and ECM rigidity—determine the effectiveness of cell migration through the dense ECM. The cell can migrate persistently even if its cortical contraction cannot deform a near-rigid ECM, but then the contraction of the cortex has to be able to sufficiently deform the nucleus. The cell can also migrate even if it fails to deform a stiff nucleus, but then it has to be able to sufficiently deform the ECM. Simulation results show that nuclear stiffness limits the cell migration more than the ECM rigidity. Simulations show the rear-squeezing mechanism of motility results in more robust migration with larger cell displacements than those with the push-pull mechanism over a range of parameter values. Additionally, results show that the rear-squeezing mechanism is aided by hydrodynamics through a pressure gradient. Computational simulations of two different mechanisms of 3D cell migration in an extracellular matrix are presented. One mechanism represents a mesenchymal mode, characterized by finger-like actin protrusions, while the second mode is more amoeboid in that rear contraction of the cortex propels the cell forward. In both mechanisms, the cell generates a thin actin protrusion on the cortex that attaches to an ECM node. The cell is then either pulled (mesenchymal) or pushed (amoeboid) forward. Results show both mechanisms result in successful migration over a range of simulated parameter values as long as the contractile tension of the cortex exceeds either the nuclear stiffness or ECM stiffness, but not necessarily both. However, the distance traveled by the amoeboid migration mode is more robust to changes in parameter values, and is larger than in simulations of the mesenchymal mode. Additionally, cells experience a favorable fluid pressure gradient when migrating in the amoeboid mode, and an adverse fluid pressure gradient in the mesenchymal mode.
Collapse
Affiliation(s)
- Ondrej Maxian
- Courant Institute of Mathematical Sciences, New York University, New York, New York, United States of America
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, New York, New York, United States of America
- Department of Biology, New York University, New York, New York, United States of America
| | - Wanda Strychalski
- Department of Mathematics, Applied Mathematics and Statistics, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
40
|
Chu HY, Chen YJ, Hsu CJ, Liu YW, Chiou JF, Lu LS, Tseng FG. Physical Cues in the Microenvironment Regulate Stemness-Dependent Homing of Breast Cancer Cells. Cancers (Basel) 2020; 12:E2176. [PMID: 32764400 PMCID: PMC7464848 DOI: 10.3390/cancers12082176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/03/2020] [Accepted: 08/04/2020] [Indexed: 12/24/2022] Open
Abstract
Tissue-specific microenvironmental factors contribute to the targeting preferences of metastatic cancers. However, the physical attributes of the premetastatic microenvironment are not yet fully characterized. In this research, we develop a transwell-based alginate hydrogel (TAH) model to study how permeability, stiffness, and roughness of a hanging alginate hydrogel regulate breast cancer cell homing. In this model, a layer of physically characterized alginate hydrogel is formed at the bottom of a transwell insert, which is placed into a matching culture well with an adherent monolayer of breast cancer cells. We found that breast cancer cells dissociate from the monolayer and home to the TAH for continual growth. The process is facilitated by the presence of rich serum in the upper chamber, the increased stiffness of the gel, as well as its surface roughness. This model is able to support the homing ability of MCF-7 and MDA-MB-231 cells drifting across the vertical distance in the culture medium. Cells homing to the TAH display stemness phenotype morphologically and biochemically. Taken together, these findings suggest that permeability, stiffness, and roughness are important physical factors to regulate breast cancer homing to a premetastatic microenvironment.
Collapse
Affiliation(s)
- Hsueh-Yao Chu
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 30013, Taiwan; (H.-Y.C.); (C.-J.H.); (Y.-W.L.)
| | - Yin-Ju Chen
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (Y.-J.C.); (J.-F.C.)
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Jieh Hsu
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 30013, Taiwan; (H.-Y.C.); (C.-J.H.); (Y.-W.L.)
| | - Yang-Wei Liu
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 30013, Taiwan; (H.-Y.C.); (C.-J.H.); (Y.-W.L.)
| | - Jeng-Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (Y.-J.C.); (J.-F.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Long-Sheng Lu
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (Y.-J.C.); (J.-F.C.)
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Fan-Gang Tseng
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu 30013, Taiwan; (H.-Y.C.); (C.-J.H.); (Y.-W.L.)
- Department of Engineering and System Science, Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing-Hua University, Hsinchu 30013, Taiwan
- Research Center for Applied Sciences, Academia Sinica, No. 128, Sec. 2, Academia Rd., Nankang, Taipei 11529, Taiwan
| |
Collapse
|
41
|
Tang VW. Collagen, stiffness, and adhesion: the evolutionary basis of vertebrate mechanobiology. Mol Biol Cell 2020; 31:1823-1834. [PMID: 32730166 PMCID: PMC7525820 DOI: 10.1091/mbc.e19-12-0709] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/11/2020] [Accepted: 05/28/2020] [Indexed: 01/09/2023] Open
Abstract
The emergence of collagen I in vertebrates resulted in a dramatic increase in the stiffness of the extracellular environment, supporting long-range force propagation and the development of low-compliant tissues necessary for the development of vertebrate traits including pressurized circulation and renal filtration. Vertebrates have also evolved integrins that can bind to collagens, resulting in the generation of higher tension and more efficient force transmission in the extracellular matrix. The stiffer environment provides an opportunity for the vertebrates to create new structures such as the stress fibers, new cell types such as endothelial cells, new developmental processes such as neural crest delamination, and new tissue organizations such as the blood-brain barrier. Molecular players found only in vertebrates allow the modification of conserved mechanisms as well as the design of novel strategies that can better serve the physiological needs of the vertebrates. These innovations collectively contribute to novel morphogenetic behaviors and unprecedented increases in the complexities of tissue mechanics and functions.
Collapse
Affiliation(s)
- Vivian W. Tang
- Department of Cell and Developmental Biology, University of Illinois, Urbana–Champaign, Urbana, IL 61801
| |
Collapse
|
42
|
Zanotelli MR, Chada NC, Johnson CA, Reinhart-King CA. The Physical Microenvironment of Tumors: Characterization and Clinical Impact. ACTA ACUST UNITED AC 2020. [DOI: 10.1142/s1793048020300029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The tumor microenvironment plays a critical role in tumorigenesis and metastasis. As tightly controlled extracellular matrix homeostasis is lost during tumor progression, a dysregulated extracellular matrix can significantly alter cellular phenotype and drive malignancy. Altered physical properties of the tumor microenvironment alter cancer cell behavior, limit delivery and efficacy of therapies, and correlate with tumorigenesis and patient prognosis. The physical features of the extracellular matrix during tumor progression have been characterized; however, a wide range of methods have been used between studies and cancer types resulting in a large range of reported values. Here, we discuss the significant mechanical and structural properties of the tumor microenvironment, summarizing their reported values and clinical impact across cancer type and grade. We attempt to integrate the values in the literature to identify sources of reported differences and commonalities to better understand how aberrant extracellular matrix dynamics contribute to cancer progression. An intimate understanding of altered matrix properties during malignant transformation will be crucial in effectively detecting, monitoring, and treating cancer.
Collapse
Affiliation(s)
- Matthew R. Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Weill Hall, Ithaca, NY 14583, USA
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Avenue, Nashville, TN 37235, USA
| | - Neil C. Chada
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Avenue, Nashville, TN 37235, USA
| | - C. Andrew Johnson
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Avenue, Nashville, TN 37235, USA
| | - Cynthia A. Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, 2414 Highland Avenue, Nashville, TN 37235, USA
| |
Collapse
|
43
|
Movilla N, Valero C, Borau C, García-Aznar JM. Matrix degradation regulates osteoblast protrusion dynamics and individual migration. Integr Biol (Camb) 2020; 11:404-413. [PMID: 31922533 DOI: 10.1093/intbio/zyz035] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 09/06/2019] [Accepted: 10/19/2019] [Indexed: 01/21/2023]
Abstract
Protrusions are one of the structures that cells use to sense their surrounding environment in a probing and exploratory manner as well as to communicate with other cells. In particular, osteoblasts embedded within a 3D matrix tend to originate a large number of protrusions compared to other type of cells. In this work, we study the role that mechanochemical properties of the extracellular matrix (ECM) play on the dynamics of these protrusions, namely, the regulation of the size and number of emanating structures. In addition, we also determine how the dynamics of the protrusions may lead the 3D movement of the osteoblasts. Significant differences were found in protrusion size and cell velocity, when degradation activity due to metalloproteases was blocked by means of an artificial broad-spectrum matrix metalloproteinase inhibitor, whereas stiffening of the matrix by introducing transglutaminase crosslinking, only induced slight changes in both protrusion size and cell velocity, suggesting that the ability of cells to create a path through the matrix is more critical than the matrix mechanical properties themselves. To confirm this, we developed a cell migration computational model in 3D including both the mechanical and chemical properties of the ECM as well as the protrusion mechanics, obtaining good agreement with experimental results.
Collapse
Affiliation(s)
- Nieves Movilla
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Clara Valero
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Carlos Borau
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Jose Manuel García-Aznar
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research, Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
44
|
The Mechanical Microenvironment in Breast Cancer. Cancers (Basel) 2020; 12:cancers12061452. [PMID: 32503141 PMCID: PMC7352870 DOI: 10.3390/cancers12061452] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 01/22/2023] Open
Abstract
Mechanotransduction is the interpretation of physical cues by cells through mechanosensation mechanisms that elegantly translate mechanical stimuli into biochemical signaling pathways. While mechanical stress and their resulting cellular responses occur in normal physiologic contexts, there are a variety of cancer-associated physical cues present in the tumor microenvironment that are pathological in breast cancer. Mechanistic in vitro data and in vivo evidence currently support three mechanical stressors as mechanical modifiers in breast cancer that will be the focus of this review: stiffness, interstitial fluid pressure, and solid stress. Increases in stiffness, interstitial fluid pressure, and solid stress are thought to promote malignant phenotypes in normal breast epithelial cells, as well as exacerbate malignant phenotypes in breast cancer cells.
Collapse
|
45
|
Pfisterer K, Levitt J, Lawson CD, Marsh RJ, Heddleston JM, Wait E, Ameer-Beg SM, Cox S, Parsons M. FMNL2 regulates dynamics of fascin in filopodia. J Cell Biol 2020; 219:e201906111. [PMID: 32294157 PMCID: PMC7199847 DOI: 10.1083/jcb.201906111] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/30/2019] [Accepted: 02/20/2020] [Indexed: 12/31/2022] Open
Abstract
Filopodia are peripheral F-actin-rich structures that enable cell sensing of the microenvironment. Fascin is an F-actin-bundling protein that plays a key role in stabilizing filopodia to support efficient adhesion and migration. Fascin is also highly up-regulated in human cancers, where it increases invasive cell behavior and correlates with poor patient prognosis. Previous studies have shown that fascin phosphorylation can regulate F-actin bundling, and that this modification can contribute to subcellular fascin localization and function. However, the factors that regulate fascin dynamics within filopodia remain poorly understood. In the current study, we used advanced live-cell imaging techniques and a fascin biosensor to demonstrate that fascin phosphorylation, localization, and binding to F-actin are highly dynamic and dependent on local cytoskeletal architecture in cells in both 2D and 3D environments. Fascin dynamics within filopodia are under the control of formins, and in particular FMNL2, that binds directly to dephosphorylated fascin. Our data provide new insight into control of fascin dynamics at the nanoscale and into the mechanisms governing rapid cytoskeletal adaptation to environmental changes. This filopodia-driven exploration stage may represent an essential regulatory step in the transition from static to migrating cancer cells.
Collapse
Affiliation(s)
- Karin Pfisterer
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - James Levitt
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
- Microscopy Innovation Centre, King's College London, Guy's Campus, London, UK
| | - Campbell D. Lawson
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Richard J. Marsh
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - John M. Heddleston
- Advanced Imaging Centre, Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | - Eric Wait
- Advanced Imaging Centre, Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | - Simon Morris Ameer-Beg
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Campus, London, UK
| | - Susan Cox
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Maddy Parsons
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| |
Collapse
|
46
|
Nerger BA, Nelson CM. Engineered extracellular matrices: emerging strategies for decoupling structural and molecular signals that regulate epithelial branching morphogenesis. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 13:103-112. [PMID: 32864528 PMCID: PMC7451493 DOI: 10.1016/j.cobme.2019.12.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The extracellular matrix (ECM) is a heterogeneous mixture of proteoglycans and fibrous proteins that form the non-cellular component of tissues and organs. During normal development, homeostasis, and disease progression, the ECM provides dynamic structural and molecular signals that influence the form and function of individual cells and multicellular tissues. Here, we review recent developments in the design and fabrication of engineered ECMs and the application of these systems to study the morphogenesis of epithelial tissues. We emphasize emerging techniques for reproducing the structural and molecular complexity of native ECM, and we highlight how these techniques may be used to decouple the different signals that drive epithelial morphogenesis. Engineered models of native ECM will enable further investigation of the dynamic mechanisms by which the microenvironment influences tissue morphogenesis.
Collapse
Affiliation(s)
- Bryan A. Nerger
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544
| | - Celeste M. Nelson
- Department of Chemical & Biological Engineering, Princeton University, Princeton, NJ 08544
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| |
Collapse
|
47
|
Abstract
The gut is a continuously renewing organ, with cell proliferation, migration, and death occurring rapidly under basal conditions. As the impact of critical illness on cell movement from crypt base to villus tip is poorly understood, the purpose of this study was to determine how sepsis alters enterocyte migration. Wild-type, transgenic, and knockout mice were injected with 5-bromo-2'deoxyuridine (BrdU) to label cells in S-phase before and after the onset of cecal ligation and puncture and were sacrificed at predetermined endpoints to determine distance proliferating cells migrated up the crypt-villus unit. Enterocyte migration rate was decreased from 24 to 96 h after sepsis. BrdU was not detectable on villi 6 days after sham laparotomy, meaning all cells had migrated the length of the gut and been exfoliated into its lumen. However, BrdU positive cells were detectable on villi 10 days after sepsis. Multiple components of gut integrity altered enterocyte migration. Sepsis decreased crypt proliferation, which further slowed enterocyte transit as mice injected with BrdU after the onset of sepsis (decreased proliferation) had slower migration than mice injected with BrdU before the onset of sepsis (normal proliferation). Decreasing intestinal apoptosis via gut-specific overexpression of Bcl-2 prevented sepsis-induced slowing of enterocyte migration. In contrast, worsened intestinal hyperpermeability by genetic deletion of JAM-A increased enterocyte migration. Sepsis therefore significantly slows enterocyte migration, and intestinal proliferation, apoptosis and permeability all affect migration time, which can potentially be targeted both genetically and pharmacologically.
Collapse
|
48
|
Avendano A, Chang JJ, Cortes-Medina MG, Seibel AJ, Admasu BR, Boutelle CM, Bushman AR, Garg AA, DeShetler CM, Cole SL, Song JW. Integrated Biophysical Characterization of Fibrillar Collagen-Based Hydrogels. ACS Biomater Sci Eng 2020; 6:1408-1417. [PMID: 32292818 DOI: 10.1021/acsbiomaterials.9b01873] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This paper describes an experimental characterization scheme of the biophysical properties of reconstituted hydrogel matrices based on indentation testing, quantification of transport via microfluidics, and confocal reflectance microscopy analysis. While methods for characterizing hydrogels exist and are widely used, they often do not measure diffusive and convective transport concurrently, determine the relationship between microstructure and transport properties, and decouple matrix mechanics and transport properties. Our integrated approach enabled independent and quantitative measurements of the structural, mechanical, and transport properties of hydrogels in a single study. We used fibrillar type I collagen as the base matrix and investigated the effects of two different matrix modifications: (1) cross-linking with human recombinant tissue transglutaminase II (hrTGII) and (2) supplementation with the nonfibrillar matrix constituent hyaluronic acid (HA). hrTGII modified the matrix structure and transport but not mechanical parameters. Furthermore, changes in the matrix structure due to hrTGII were seen to be dependent on the concentration of collagen. In contrast, supplementation of HA at different collagen concentrations altered the matrix microstructure and mechanical indentation behavior but not transport parameters. These experimental observations reveal the important relationship between extracellular matrix (ECM) composition and biophysical properties. The integrated techniques are versatile, robust, and accessible; and as matrix-cell interactions are instrumental for many biological processes, the methods and findings described here should be broadly applicable for characterizing hydrogel materials used for three-dimensional (3-D) tissue-engineered culture models.
Collapse
Affiliation(s)
- Alex Avendano
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jonathan J Chang
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Marcos G Cortes-Medina
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Aaron J Seibel
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Bitania R Admasu
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Cassandra M Boutelle
- Department of Integrated Systems Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Andrew R Bushman
- William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | - Ayush Arpit Garg
- Department of Biomedical Engineering and Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, Ohio 43210, United States
| | | | - Sara L Cole
- Campus Microscopy and Imaging Facility, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jonathan W Song
- Department of Mechanical and Aerospace Engineering and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
49
|
Davidson MD, Ban E, Schoonen ACM, Lee MH, D'Este M, Shenoy VB, Burdick JA. Mechanochemical Adhesion and Plasticity in Multifiber Hydrogel Networks. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1905719. [PMID: 31851400 PMCID: PMC7042082 DOI: 10.1002/adma.201905719] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/14/2019] [Indexed: 05/27/2023]
Abstract
The extracellular matrix (ECM) has force-responsive (i.e., mechanochemical) properties that enable adaptation to mechanical loading through changes in fibrous network structure and interfiber bonding. Imparting such properties into synthetic fibrous materials will allow reinforcement under mechanical load, the potential for material self-adhesion, and the general mimicking of ECM. Multifiber hydrogel networks are developed through the electrospinning of multiple fibrous hydrogel populations, where fibers contain complementary chemical moieties (e.g., aldehyde and hydrazide groups) that form covalent bonds within minutes when brought into contact under mechanical load. These fiber interactions lead to microscale anisotropy, as well as increased material stiffness and plastic deformation. Macroscale structures (e.g., tubes and layered scaffolds) are fabricated from these materials through interfiber bonding and adhesion when placed into contact while maintaining a microscale fibrous architecture. The design principles for engineering plasticity described can be applied to numerous material systems to introduce unique properties, from textiles to biomedical applications.
Collapse
Affiliation(s)
- Matthew D Davidson
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ehsan Ban
- Center for Engineering Mechanobiology and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anna C M Schoonen
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mu-Huan Lee
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Matteo D'Este
- AO Research Institute Davos, 7270, Davos, Switzerland
| | - Vivek B Shenoy
- Center for Engineering Mechanobiology and Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
50
|
Konar S, Edwina P, Ramanujam V, Arunachalakasi A, Bajpai SK. Collagen-I/silk-fibroin biocomposite exhibits microscalar confinement of cells and induces anisotropic morphology and migration of embedded fibroblasts. J Biomed Mater Res B Appl Biomater 2020; 108:2368-2377. [PMID: 31984672 DOI: 10.1002/jbm.b.34570] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/06/2019] [Accepted: 01/13/2020] [Indexed: 12/17/2022]
Abstract
Microstructural anisotropy of tumor-associated matrix correlates with invasion of cancer cells into the surrounding matrix during metastasis. Here, we report the fabrication and characterization of a three-dimensional (3D) silk-fibroin/collagen-I bio-composite based cell-culture model that exhibits microstructural and biochemical anisotropy. Using RGD-deficient silk-fibroin fibers to confine collagen-I gelation, we develop a silk-fibroin/collagen-I (SFC) bio-composite in a one-step process allowing control over the microstructural and biochemical anisotropy and the pore-size. Two forms of the SFC bio-composite are reported: a sandwich (Sfc ) configuration amenable to live-cell microscopy and an unsupported membrane (Mfc ) for use as a scaffold. Both microscalar and macroscalar mechanical properties of the SFC bio-composite are characterized using atomic force microscope (AFM)-based indentation and tensile-testing. We find that the modulus of stiffness of both Sfc and Mfc can be controlled and falls in the physiological range of 5-20 kPa. Furthermore, the modulus of stiffness of Mfc exhibits a ~200% increase in axial direction of microstructure, as compared to lateral direction. This implies a highly anisotropic mechanical stiffness of the microenvironment. Live-cell morphology and migration studies show that both the morphology and the migration of NIH-3 T3 fibroblasts is anisotropic and correlates with microstructural anisotropy. Our results show that SFC bio-composite permits proliferation of cells in both Sfc and Mfc configuration, promotes cell-migration along the major axis of anisotropy and together with morphological and migration data, suggest a potential application of both the composite configurations as a biomimetic scaffold for tissue engineering applications.
Collapse
Affiliation(s)
- Subhajit Konar
- Department of Applied Mechanics, Indian Institute of Technology - Madras, Chennai, India.,Faculty of Medical and Health Sciences, Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Privita Edwina
- Department of Applied Mechanics, Indian Institute of Technology - Madras, Chennai, India
| | - Vaibavi Ramanujam
- Department of Applied Mechanics, Indian Institute of Technology - Madras, Chennai, India
| | | | - Saumendra Kumar Bajpai
- Department of Applied Mechanics, Indian Institute of Technology - Madras, Chennai, India
| |
Collapse
|