51
|
Chen MB, Kamm RD, Moeendarbary E. Engineered Models of Metastasis with Application to Study Cancer Biomechanics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1092:189-207. [PMID: 30368754 DOI: 10.1007/978-3-319-95294-9_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Three-dimensional complex biomechanical interactions occur from the initial steps of tumor formation to the later phases of cancer metastasis. Conventional monolayer cultures cannot recapitulate the complex microenvironment and chemical and mechanical cues that tumor cells experience during their metastatic journey, nor the complexity of their interactions with other, noncancerous cells. As alternative approaches, various engineered models have been developed to recapitulate specific features of each step of metastasis with tunable microenvironments to test a variety of mechanistic hypotheses. Here the main recent advances in the technologies that provide deeper insight into the process of cancer dissemination are discussed, with an emphasis on three-dimensional and mechanical factors as well as interactions between multiple cell types.
Collapse
Affiliation(s)
- Michelle B Chen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Emad Moeendarbary
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, University College London, London, UK
| |
Collapse
|
52
|
Chen BJ, Tang YJ, Tang YL, Liang XH. What makes cells move: Requirements and obstacles for leader cells in collective invasion. Exp Cell Res 2019; 382:111481. [PMID: 31247191 DOI: 10.1016/j.yexcr.2019.06.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/15/2019] [Accepted: 06/23/2019] [Indexed: 02/05/2023]
Abstract
Most recently, mounting evidence has shown that cancer cells can invade as a cohesive and multicellular group with coordinated movement, which is called collective invasion. In this cohesive cancer cell group, cancer cells at the front of collective invasion are defined as leader cell that are responsible for many aspects of collective invasion, including sensing the microenvironment, determining the invasion direction, modifying the path of invasion and transmitting information to other cells. To fulfill their dispensable roles, leader cells are required to embark on some specific phenotypes with unusual expression of some proteins and it's very important to investigate into these proteins as they may serve as potential therapeutic targets. Here, in this review we will summarize current knowledge on four emerging proteins highly expressed in leader cells including K14, ΔNp63α, Dll4 and cysteine protease cathepsin B (CTSB), with a focus on their important roles in collective invasion and special mechanisms by which they promote collective invasion.
Collapse
Affiliation(s)
- Bing-Jun Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, China.
| | - Ya-Jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China.
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral Pathology, West China Hospital of Stomatology, Sichuan University.China.
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, China.
| |
Collapse
|
53
|
Evje S, Waldeland JO. How Tumor Cells Can Make Use of Interstitial Fluid Flow in a Strategy for Metastasis. Cell Mol Bioeng 2019; 12:227-254. [PMID: 31719912 DOI: 10.1007/s12195-019-00569-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/19/2019] [Indexed: 12/18/2022] Open
Abstract
Introduction The phenomenon of lymph node metastasis has been known for a long time. However, the underlying mechanism by which malignant tumor cells are able to break loose from the primary tumor site remains unclear. In particular, two competing fluid sensitive migration mechanisms have been reported in the experimental literature: (i) autologous chemotaxis (Shields et al. in Cancer Cell 11:526-538, 2007) which gives rise to downstream migration; (ii) an integrin-mediated and strain-induced upstream mechanism (Polacheck et al. in PNAS 108:11115-11120, 2011). How can these two competing mechanisms be used as a means for metastatic behavior in a realistic tumor setting? Excessive fluid flow is typically produced from leaky intratumoral blood vessels and collected by lymphatics in the peritumoral region giving rise to a heterogeneous fluid velocity field and a corresponding heterogeneous cell migration behavior, quite different from the experimental setup. Method In order to shed light on this issue there is a need for tools which allow one to extrapolate the observed single cell behavior in a homogeneous microfluidic environment to a more realistic, higher-dimensional tumor setting. Here we explore this issue by using a computational multiphase model. The model has been trained with data from the experimental results mentioned above which essentially reflect one-dimensional behavior. We extend the model to an envisioned idealized two-dimensional tumor setting. Result A main observation from the simulation is that the autologous chemotaxis migration mechanism, which triggers tumor cells to go with the flow in the direction of lymphatics, becomes much more aggressive and effective as a means for metastasis in the presence of realistic IF flow. This is because the outwardly directed IF flow generates upstream cell migration that possibly empowers small clusters of tumor cells to break loose from the primary tumor periphery. Without this upstream stress-mediated migration, autologous chemotaxis is inclined to move cells at the rim of the tumor in a homogeneous and collective, but space-demanding style. In contrast, inclusion of realistic IF flow generates upstream migration that allows two different aspects to be synthesized: maintain the coherency and solidity of the the primary tumor and at the same time cleave the outgoing waves of tumor cells into small clusters at the front that can move collectively in a more specific direction.
Collapse
Affiliation(s)
- Steinar Evje
- Department of Energy and Petroleum, University of Stavanger, 4068 Stavanger, Norway
| | - Jahn Otto Waldeland
- Department of Energy and Petroleum, University of Stavanger, 4068 Stavanger, Norway
| |
Collapse
|
54
|
Elhebeary M, Emon MAB, Aydin O, Saif MTA. A novel technique for in situ uniaxial tests of self-assembled soft biomaterials. LAB ON A CHIP 2019; 19:1153-1161. [PMID: 30776038 PMCID: PMC6437030 DOI: 10.1039/c8lc01273c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
We introduce a novel method to form 3D biomimetic tissues from a droplet of a cell-extracellular matrix (ECM) mixture on a sensor stage and to quantify tissue force and stiffness as a function of time under optical microscopes. This method exploits advances in micro-nano fabrication and capillarity for self-assembly and self-alignment of tissues on the stage. It allows simultaneous investigation of the microstructure of the tissue in situ while its mechanical response is quantified, thus linking tissue biophysics with physiology and revealing structural-functional properties of 3D tissues. We demonstrate the functionality of the stage by studying the mechanical behavior of different cell-collagen mixtures under mechanical, chemical and electrical stimulation. This includes force evolution in cell-free collagen during curing, myotubes differentiated from muscle cell-collagen/Matrigel ECM subjected to electrical stimulation, and fibroblast-collagen tissue subjected to cancer cell conditioned media (CM) and a Rho-kinase inhibitor, Y27632. Muscle contraction decreases with increasing frequency of electrical stimulation, and fibroblasts respond to CM by increasing contractility for a short time and completely relax in the presence of Y27632 but restore force with Y27632 washout.
Collapse
Affiliation(s)
- Mohamed Elhebeary
- University of Illinois at Urbana-Champaign, 1206 W. Green St, Urbana, IL 61801, USA.
| | | | | | | |
Collapse
|
55
|
Zhang L, Hao C, Wu Y, Zhu Y, Ren Y, Tong Z. Microcalcification and BMP-2 in breast cancer: correlation with clinicopathological features and outcomes. Onco Targets Ther 2019; 12:2023-2033. [PMID: 30936719 PMCID: PMC6421899 DOI: 10.2147/ott.s187835] [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] [Indexed: 12/20/2022] Open
Abstract
Background Microcalcification is a very important diagnostic information in breast cancer. The purpose of this study was to determine the relationship of clinicopathological features and prognosis of breast cancer with microcalcification and to detect biomarkers related to the possible mechanisms of microcalcifications. Patients and methods All 529 subjects with microcalcifications were selected from patients who had been examined using breast mammography. The control group did not have detectable microcalcifications, and was matched in a ratio of 1:3. The clinicopathological factors, progression-free survival (PFS), and overall survival were evaluated by SPSS. Results There was a significant difference in tumor size between the two groups, with larger tumors in the calcification group than the control group, and the proportion of patients in the calcification group with tumors of >5 cm was 20.4% vs 17.2% in the control group (P=0.041). The proportion of patients with lymph node metastasis in the calcification group was higher than that of the control group (35% vs 27.9%, P=0.027). The recurrence rate in ductal carcinoma in situ (DCIS) and invasive ductal carcinoma (IDC) patients with microcalcification was higher than that in the control group (P=0.035 and 0.044). BMP-2 expression was higher in breast cancer tissues, especially in breast cancer tissues with microcalcifications. The recurrence rate in the BMP-2(+) group was higher than that in the BMP-2(-) group both in DCIS and IDC (P=0.044 and 0.049). Microcalcifications and the positive expression of BMP-2 were independent factors affecting the PFS of the breast cancer patients. Conclusion Through the analysis of this study, it was found that the prognosis of the patients with microcalcification was relatively poor. BMP-2 was highly expressed in the breast cancer with microcalcification and was associated with poor prognosis.
Collapse
Affiliation(s)
- Li Zhang
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| | - Chunfang Hao
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| | - Yansheng Wu
- Department of Maxillofacial and Otorhinolaryngology Head and Neck Surgery, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Yuying Zhu
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| | - Yulin Ren
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| | - Zhongsheng Tong
- Department of Breast Oncology, Key Laboratory of Breast Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China,
| |
Collapse
|
56
|
Smeland HYH, Lu N, Karlsen TV, Salvesen G, Reed RK, Stuhr L. Stromal integrin α11-deficiency reduces interstitial fluid pressure and perturbs collagen structure in triple-negative breast xenograft tumors. BMC Cancer 2019; 19:234. [PMID: 30876468 PMCID: PMC6419843 DOI: 10.1186/s12885-019-5449-z] [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: 06/01/2018] [Accepted: 03/10/2019] [Indexed: 12/16/2022] Open
Abstract
Background Cancer progression is influenced by a pro-tumorigenic microenvironment. The aberrant tumor stroma with increased collagen deposition, contractile fibroblasts and dysfunctional vessels has a major impact on the interstitial fluid pressure (PIF) in most solid tumors. An increased tumor PIF is a barrier to the transport of interstitial fluid into and within the tumor. Therefore, understanding the mechanisms that regulate pressure homeostasis can lead to new insight into breast tumor progression, invasion and response to therapy. The collagen binding integrin α11β1 is upregulated during myofibroblast differentiation and expressed on fibroblasts in the tumor stroma. As a collagen organizer and a probable link between contractile fibroblasts and the complex collagen network in tumors, integrin α11β1 could be a potential regulator of tumor PIF. Methods We investigated the effect of stromal integrin α11-deficiency on pressure homeostasis, collagen organization and tumor growth using orthotopic and ectopic triple-negative breast cancer xenografts (MDA-MB-231 and MDA-MB-468) in wild type and integrin α11-deficient mice. PIF was measured by the wick-in-needle technique, collagen by Picrosirius Red staining and electron microscopy, and uptake of radioactively labeled 5FU by microdialysis. Further, PIF in heterospheroids composed of MDA-MB-231 cells and wild type or integrin α11-deficient fibroblasts was measured by micropuncture. Results Stromal integrin α11-deficiency decreased PIF in both the orthotopic breast cancer models. A concomitant perturbed collagen structure was seen, with fewer aligned and thinner fibrils. Integrin α11-deficiency also impeded MDA-MB-231 breast tumor growth, but no effect was observed on drug uptake. No effects were seen in the ectopic model. By investigating the isolated effect of integrin α11-positive fibroblasts on MDA-MB-231 cells in vitro, we provide evidence that PIF regulation was mediated by integrin α11-positive fibroblasts. Conclusion We hereby show the importance of integrin α11β1 in pressure homeostasis in triple-negative breast tumors, indicating a new role for integrin α11β1 in the tumor microenvironment. Our data suggest that integrin α11β1 has a pro-tumorigenic effect on triple-negative breast cancer growth in vivo. The significance of the local microenvironment is shown by the different effects of integrin α11β1 in the orthotopic and ectopic models, underlining the importance of choosing an appropriate preclinical model. Electronic supplementary material The online version of this article (10.1186/s12885-019-5449-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hilde Ytre-Hauge Smeland
- Department of Biomedicine, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway. .,Centre of Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway.
| | - Ning Lu
- Department of Biomedicine, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway.,Centre of Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway
| | - Tine V Karlsen
- Department of Biomedicine, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway
| | - Gerd Salvesen
- Department of Biomedicine, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway
| | - Rolf K Reed
- Department of Biomedicine, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway.,Centre of Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway
| | - Linda Stuhr
- Department of Biomedicine, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway.,Centre of Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, P.O. Box 7804, 5020, Bergen, Norway
| |
Collapse
|
57
|
Hwang PY, Brenot A, King AC, Longmore GD, George SC. Randomly Distributed K14 + Breast Tumor Cells Polarize to the Leading Edge and Guide Collective Migration in Response to Chemical and Mechanical Environmental Cues. Cancer Res 2019; 79:1899-1912. [PMID: 30862718 DOI: 10.1158/0008-5472.can-18-2828] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/27/2018] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Collective cell migration is an adaptive, coordinated interactive process involving cell-cell and cell-extracellular matrix (ECM) microenvironmental interactions. A critical aspect of collective migration is the sensing and establishment of directional movement. It has been proposed that a subgroup of cells known as leader cells localize at the front edge of a collectively migrating cluster and are responsible for directing migration. However, it is unknown how and when leader cells arrive at the front edge and what environmental cues dictate leader cell development and behavior. Here, we addressed these questions by combining a microfluidic device design that mimics multiple tumor microenvironmental cues concurrently with biologically relevant primary, heterogeneous tumor cell organoids. Prior to migration, breast tumor leader cells (K14+) were present throughout a tumor organoid and migrated (polarized) to the leading edge in response to biochemical and biomechanical cues. Impairment of either CXCR4 (biochemical responsive) or the collagen receptor DDR2 (biomechanical responsive) abrogated polarization of leader cells and directed collective migration. This work demonstrates that K14+ leader cells utilize both chemical and mechanical cues from the microenvironment to polarize to the leading edge of collectively migrating tumors. SIGNIFICANCE: These findings demonstrate that pre-existing, randomly distributed leader cells within primary tumor organoids use CXCR4 and DDR2 to polarize to the leading edge and direct migration.
Collapse
Affiliation(s)
- Priscilla Y Hwang
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri.,ICCE Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Audrey Brenot
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri.,ICCE Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Ashley C King
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri.,ICCE Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Gregory D Longmore
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri. .,ICCE Institute, Washington University in St. Louis, St. Louis, Missouri.,Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, Missouri
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, California.
| |
Collapse
|
58
|
Shang M, Soon RH, Lim CT, Khoo BL, Han J. Microfluidic modelling of the tumor microenvironment for anti-cancer drug development. LAB ON A CHIP 2019; 19:369-386. [PMID: 30644496 DOI: 10.1039/c8lc00970h] [Citation(s) in RCA: 152] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Cancer is the leading cause of death worldwide. The complex and disorganized tumor microenvironment makes it very difficult to treat this disease. The most common in vitro drug screening method now is based on 2D culture models which poorly represent actual tumors. Therefore, many 3D tumor models which are more physiologically relevant have been developed to conduct in vitro drug screening and alleviate this situation. Among all these models, the microfluidic tumor model has the unique advantage of recapitulating the tumor microenvironment in a comparatively easier and representative fashion. While there are many review papers available on the related topic of microfluidic tumor models, in this review we aim to focus more on the possibility of generating "clinically actionable information" from these microfluidic systems, besides scientific insight. Our topics cover the tumor microenvironment, conventional 2D and 3D cultures, animal models, and microfluidic tumor models, emphasizing their link to anti-cancer drug discovery and personalized medicine.
Collapse
Affiliation(s)
- Menglin Shang
- BioSystems and Micromechanics (BioSyM) IRG, Singapore-MIT Alliance for Research and Technology (SMART) Centre, 1, Create Way, Enterprise Wing, 138602, Singapore.
| | | | | | | | | |
Collapse
|
59
|
Yeldag G, Rice A, Del Río Hernández A. Chemoresistance and the Self-Maintaining Tumor Microenvironment. Cancers (Basel) 2018; 10:E471. [PMID: 30487436 PMCID: PMC6315745 DOI: 10.3390/cancers10120471] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
The progression of cancer is associated with alterations in the tumor microenvironment, including changes in extracellular matrix (ECM) composition, matrix rigidity, hypervascularization, hypoxia, and paracrine factors. One key malignant phenotype of cancer cells is their ability to resist chemotherapeutics, and elements of the ECM can promote chemoresistance in cancer cells through a variety of signaling pathways, inducing changes in gene expression and protein activity that allow resistance. Furthermore, the ECM is maintained as an environment that facilitates chemoresistance, since its constitution modulates the phenotype of cancer-associated cells, which themselves affect the microenvironment. In this review, we discuss how the properties of the tumor microenvironment promote chemoresistance in cancer cells, and the interplay between these external stimuli. We focus on both the response of cancer cells to the external environment, as well as the maintenance of the external environment, and how a chemoresistant phenotype emerges from the complex signaling network present.
Collapse
Affiliation(s)
- Gulcen Yeldag
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| | - Armando Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College London, London, UK.
| |
Collapse
|
60
|
Chu Y, Fang Y, Chi J, Li J, Zhang D, Zou Y, Wang Z. Astragalus polysaccharides decrease proliferation, migration, and invasion but increase apoptosis of human osteosarcoma cells by up-regulation of microRNA-133a. ACTA ACUST UNITED AC 2018; 51:e7665. [PMID: 30462772 PMCID: PMC6247244 DOI: 10.1590/1414-431x20187665] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 09/12/2018] [Indexed: 11/21/2022]
Abstract
Osteosarcoma (OS) has a high incidence, malignity, and frequency of recurrence and metastasis. In this study, we aimed to explore the potential anti-cancer effects of Astragalus polysaccharides (APS) on human OS MG63 cells as well as underlying mechanisms. Viability of MG63 cells was assessed by CCK-8 assay to determine the adequate concentration of APS. Then, effects of APS on MG63 cell proliferation, cell cycle distribution, apoptosis, and migration and invasion were analyzed by BrdU incorporation, PI staining, flow cytometry, and transwell assays, respectively. The expression levels of proteins involved in these physiological processes were assessed by western blot analysis. Afterwards, miR-133a level in APS-treated cells was determined by qRT-PCR, and whether APS affected MG63 cells through regulation of miR-133a was determined. Finally, the activation of c-Jun N-terminal protein kinase (JNK) pathway was detected. We found that APS treatment suppressed the viability, proliferation, migration, and invasion of MG63 cells, as well as induced cell apoptosis. Moreover, APS enhanced the expression of miR-133a in MG63 cells. Knockdown of miR-133a reversed the APS treatment-induced MG63 cell proliferation, migration and invasion inhibition, as well as cell apoptosis. Furthermore, APS inactivated JNK pathway in MG63 cells. Knockdown of miR-133a reversed the APS treatment-induced inactivation of JNK pathway in MG63 cells. To conclude, APS repressed proliferation, migration, and invasion while induced apoptosis of OS MG63 cells by up-regulating miR-133a and then inactivating JNK pathway.
Collapse
Affiliation(s)
- Yanchen Chu
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yuan Fang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jingwei Chi
- Key Laboratory of Thyroidism, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jing Li
- Department of Gynaecology and Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Dongyang Zhang
- Department of Orthopedics, Laixi People's Hospital, Laixi, Shandong, China
| | - Yunwen Zou
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Zhijie Wang
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
61
|
FOXF2 inhibits proliferation, migration, and invasion of Hela cells by regulating Wnt signaling pathway. Biosci Rep 2018; 38:BSR20180747. [PMID: 30249755 PMCID: PMC6200704 DOI: 10.1042/bsr20180747] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 09/02/2018] [Accepted: 09/17/2018] [Indexed: 12/19/2022] Open
Abstract
This article was aimed to study the FOXF2 effects on cervical cancer. Tumor tissues and adjacent tissues of 41 cervical cancer patients were collected. Human endometrial epithelial cells (hEEC) and Hela cells were cultured. FOXF2 expression vector and its empty vector were transfected into Hela cells, and named as pcDNA 3.1-FOXF2 group and Vector group, respectively. Hela cells without any treatment were set as Blank group. qRT-PCR was used to detect mRNA expression. Nude mouse xenograft assay was performed to test Hela cells proliferation ability in vivo. FOXF2 and β-catenin positive cell numbers were detected by immunohistochemistry. Protein expression was analyzed by Western blot. Cells migration and invasion were conducted by Transwell. Tumor tissues and Hela cells FOXF2 expression were lower than that in adjacent tissues and hEEC (P<0.01). Low FOXF2 expression predicted poor outcomes of cervical cancer patients. Compared with Blank group and Vector group, Hela cells of pcDNA 3.1-FOXF2 group were with higher FOXF2 expression, lower OD495 value, migrated and invaded cells, higher E-cadherin expression, lower Vimentin and Snail expression, smaller tumor volume in nude mice, lower c-Myc, CyclinDl, MMP9, Lgr5, and nuclear β-catenin expression (all P<0.01). FOXF2 inhibits Hela cells proliferation, migration, and invasion through regulating Wnt signaling pathway.
Collapse
|
62
|
Klymenko Y, Wates RB, Weiss-Bilka H, Lombard R, Liu Y, Campbell L, Kim O, Wagner D, Ravosa MJ, Stack MS. Modeling the effect of ascites-induced compression on ovarian cancer multicellular aggregates. Dis Model Mech 2018; 11:dmm034199. [PMID: 30254133 PMCID: PMC6176988 DOI: 10.1242/dmm.034199] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/29/2018] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy. EOC dissemination is predominantly via direct extension of cells and multicellular aggregates (MCAs) into the peritoneal cavity, which adhere to and induce retraction of peritoneal mesothelium and proliferate in the submesothelial matrix to generate metastatic lesions. Metastasis is facilitated by the accumulation of malignant ascites (500 ml to >2 l), resulting in physical discomfort and abdominal distension, and leading to poor prognosis. Although intraperitoneal fluid pressure is normally subatmospheric, an average intraperitoneal pressure of 30 cmH2O (22.1 mmHg) has been reported in women with EOC. In this study, to enable experimental evaluation of the impact of high intraperitoneal pressure on EOC progression, two new in vitro model systems were developed. Initial experiments evaluated EOC MCAs in pressure vessels connected to an Instron to apply short-term compressive force. A Flexcell Compression Plus system was then used to enable longer-term compression of MCAs in custom-designed hydrogel carriers. Results show changes in the expression of genes related to epithelial-mesenchymal transition as well as altered dispersal of compressed MCAs on collagen gels. These new model systems have utility for future analyses of compression-induced mechanotransduction and the resulting impact on cellular responses related to intraperitoneal metastatic dissemination.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Yuliya Klymenko
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Rebecca B Wates
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Holly Weiss-Bilka
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel Lombard
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Yueying Liu
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Leigh Campbell
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Oleg Kim
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Mathematics, University of California, Riverside, CA 92521, USA
| | - Diane Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Matthew J Ravosa
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - M Sharon Stack
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
63
|
Extracellular fluid viscosity enhances liver cancer cell mechanosensing and migration. Biomaterials 2018; 177:113-124. [DOI: 10.1016/j.biomaterials.2018.05.058] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/10/2018] [Accepted: 05/31/2018] [Indexed: 12/31/2022]
|
64
|
Sleeboom JJF, Eslami Amirabadi H, Nair P, Sahlgren CM, den Toonder JMJ. Metastasis in context: modeling the tumor microenvironment with cancer-on-a-chip approaches. Dis Model Mech 2018; 11:11/3/dmm033100. [PMID: 29555848 PMCID: PMC5897732 DOI: 10.1242/dmm.033100] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Most cancer deaths are not caused by the primary tumor, but by secondary tumors formed through metastasis, a complex and poorly understood process. Cues from the tumor microenvironment, such as the biochemical composition, cellular population, extracellular matrix, and tissue (fluid) mechanics, have been indicated to play a pivotal role in the onset of metastasis. Dissecting the role of these cues from the tumor microenvironment in a controlled manner is challenging, but essential to understanding metastasis. Recently, cancer-on-a-chip models have emerged as a tool to study the tumor microenvironment and its role in metastasis. These models are based on microfluidic chips and contain small chambers for cell culture, enabling control over local gradients, fluid flow, tissue mechanics, and composition of the local environment. Here, we review the recent contributions of cancer-on-a-chip models to our understanding of the role of the tumor microenvironment in the onset of metastasis, and provide an outlook for future applications of this emerging technology. Summary: This Review evaluates the recent contributions of cancer-on-a-chip models to our understanding of the tumor microenvironment and its role in the onset of metastasis. The authors also provide an outlook for future applications of this emerging technology.
Collapse
Affiliation(s)
- Jelle J F Sleeboom
- Microsystems Group, Department of Mechanical Engineering, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands.,Soft Tissue Engineering & Mechanobiology, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands
| | - Hossein Eslami Amirabadi
- Microsystems Group, Department of Mechanical Engineering, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands
| | - Poornima Nair
- Microsystems Group, Department of Mechanical Engineering, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands
| | - Cecilia M Sahlgren
- Soft Tissue Engineering & Mechanobiology, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands.,Turku Centre for Biotechnology, Åbo Akademi University, Domkyrkotorget 3, FI-20500, Turku, Finland
| | - Jaap M J den Toonder
- Microsystems Group, Department of Mechanical Engineering, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands .,Institute for Complex Molecular Systems, Eindhoven University of Technology, Gemini-Zuid, Groene Loper 15, 5612AZ, Eindhoven, The Netherlands
| |
Collapse
|
65
|
Silver BB, Nelson CM. The Bioelectric Code: Reprogramming Cancer and Aging From the Interface of Mechanical and Chemical Microenvironments. Front Cell Dev Biol 2018; 6:21. [PMID: 29560350 PMCID: PMC5845671 DOI: 10.3389/fcell.2018.00021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/15/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer is a complex, heterogeneous group of diseases that can develop through many routes. Broad treatments such as chemotherapy destroy healthy cells in addition to cancerous ones, but more refined strategies that target specific pathways are usually only effective for a limited number of cancer types. This is largely due to the multitude of physiological variables that differ between cells and their surroundings. It is therefore important to understand how nature coordinates these variables into concerted regulation of growth at the tissue scale. The cellular microenvironment might then be manipulated to drive cells toward a desired outcome at the tissue level. One unexpected parameter, cellular membrane voltage (Vm), has been documented to exert control over cellular behavior both in culture and in vivo. Manipulating this fundamental cellular property influences a remarkable array of organism-wide patterning events, producing striking outcomes in both tumorigenesis as well as regeneration. These studies suggest that Vm is not only a key intrinsic cellular property, but also an integral part of the microenvironment that acts in both space and time to guide cellular behavior. As a result, there is considerable interest in manipulating Vm both to treat cancer as well as to regenerate organs damaged or deteriorated during aging. However, such manipulations have produced conflicting outcomes experimentally, which poses a substantial barrier to understanding the fundamentals of bioelectrical reprogramming. Here, we summarize these inconsistencies and discuss how the mechanical microenvironment may impact bioelectric regulation.
Collapse
Affiliation(s)
- Brian B. Silver
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
| | - Celeste M. Nelson
- Department of Molecular Biology, Princeton University, Princeton, NJ, United States
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, United States
| |
Collapse
|
66
|
Huang YL, Segall JE, Wu M. Microfluidic modeling of the biophysical microenvironment in tumor cell invasion. LAB ON A CHIP 2017; 17:3221-3233. [PMID: 28805874 PMCID: PMC6007858 DOI: 10.1039/c7lc00623c] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Tumor cell invasion, whether penetrating through the extracellular matrix (ECM) or crossing a vascular endothelium, is a critical step in the cancer metastatic cascade. Along the way from a primary tumor to a distant metastatic site, tumor cells interact actively with the microenvironment either via biomechanical (e. g. ECM stiffness) or biochemical (e.g. secreted cytokines) signals. Increasingly, it is recognized that the tumor microenvironment (TME) is a critical player in tumor cell invasion. A main challenge for the mechanistic understanding of tumor cell-TME interactions comes from the complexity of the TME, which consists of extracellular matrices, fluid flows, cytokine gradients and other cell types. It is difficult to control TME parameters in conventional in vitro experimental designs such as Boyden chambers or in vivo such as in mouse models. Microfluidics has emerged as an enabling tool for exploring the TME parameter space because of its ease of use in recreating a complex and physiologically realistic three dimensional TME with well-defined spatial and temporal control. In this perspective, we will discuss designing principles for modeling the biophysical microenvironment (biological flows and ECM) for tumor cells using microfluidic devices and the potential microfluidic technology holds in recreating a physiologically realistic tumor microenvironment. The focus will be on applications of microfluidic models in tumor cell invasion.
Collapse
Affiliation(s)
- Yu Ling Huang
- Department of Biological and Environmental Engineering, Cornell University, 306 Riley-Robb Hall, Ithaca, NY 14853, USA.
| | | | | |
Collapse
|
67
|
Piotrowski-Daspit AS, Nerger BA, Wolf AE, Sundaresan S, Nelson CM. Dynamics of Tissue-Induced Alignment of Fibrous Extracellular Matrix. Biophys J 2017; 113:702-713. [PMID: 28793224 DOI: 10.1016/j.bpj.2017.06.046] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 06/14/2017] [Accepted: 06/23/2017] [Indexed: 12/21/2022] Open
Abstract
Aligned fibers of extracellular matrix (ECM) affect the direction, efficiency, and persistence of migrating cells. To uncover the mechanisms by which multicellular tissues align their surrounding ECM before migration, we used an engineered three-dimensional culture model to investigate the dynamics of ECM alignment around tissues of defined geometry. Analysis of ECM alignment over time revealed that tissues rapidly reorganize their surrounding matrix, with a characteristic time that depends on the type of cell and the initial tissue geometry. We found that matrix metalloproteinase activity is not required for matrix alignment before cell migration. Instead, alignment is driven by Rho-mediated cytoskeletal contractility and accelerated by propagation of tension through intercellular adhesions. Our data suggest that multicellular tissues align their surrounding matrix by pulling collectively to exert strain, which is primarily a physical process. Consistently, the pattern of matrix alignment depends on tissue geometry and the resulting distribution of mechanical strain, with asymmetric tissues generating a higher degree of matrix alignment along their longest axes. The rapid ability of multicellular tissues to physically remodel their matrix enables their constituent cells to migrate efficiently along aligned fibers and to quickly change their direction according to other microenvironmental cues, which is important for both normal and disease processes.
Collapse
Affiliation(s)
| | - Bryan A Nerger
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey
| | - Abraham E Wolf
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey
| | - Sankaran Sundaresan
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey; Department of Molecular Biology, Princeton University, Princeton, New Jersey.
| |
Collapse
|
68
|
Cell Division Induces and Switches Coherent Angular Motion within Bounded Cellular Collectives. Biophys J 2017; 112:2419-2427. [PMID: 28591614 DOI: 10.1016/j.bpj.2017.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/17/2017] [Accepted: 05/02/2017] [Indexed: 12/30/2022] Open
Abstract
Collective cell migration underlies many biological processes, including embryonic development, wound healing, and cancer progression. In the embryo, cells have been observed to move collectively in vortices using a mode of collective migration known as coherent angular motion (CAM). To determine how CAM arises within a population and changes over time, here, we study the motion of mammary epithelial cells within engineered monolayers, in which the cells move collectively about a central axis in the tissue. Using quantitative image analysis, we find that CAM is significantly reduced when mitosis is suppressed. Particle-based simulations recreate the observed trends, suggesting that cell divisions drive the robust emergence of CAM and facilitate switches in the direction of collective rotation. Our simulations predict that the location of a dividing cell, rather than the orientation of the division axis, facilitates the onset of this motion. These predictions agree with experimental observations, thereby providing, to our knowledge, new insight into how cell divisions influence CAM within a tissue. Overall, these findings highlight the dynamic nature of CAM and suggest that regulating cell division is crucial for tuning emergent collective migratory behaviors, such as vortical motions observed in vivo.
Collapse
|
69
|
Klymenko Y, Kim O, Stack MS. Complex Determinants of Epithelial: Mesenchymal Phenotypic Plasticity in Ovarian Cancer. Cancers (Basel) 2017; 9:cancers9080104. [PMID: 28792442 PMCID: PMC5575607 DOI: 10.3390/cancers9080104] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/02/2017] [Accepted: 08/06/2017] [Indexed: 02/07/2023] Open
Abstract
Unlike most epithelial malignancies which metastasize hematogenously, metastasis of epithelial ovarian cancer (EOC) occurs primarily via transcoelomic dissemination, characterized by exfoliation of cells from the primary tumor, avoidance of detachment-induced cell death (anoikis), movement throughout the peritoneal cavity as individual cells and multi-cellular aggregates (MCAs), adhesion to and disruption of the mesothelial lining of the peritoneum, and submesothelial matrix anchoring and proliferation to generate widely disseminated metastases. This exceptional microenvironment is highly permissive for phenotypic plasticity, enabling mesenchymal-to-epithelial (MET) and epithelial-to-mesenchymal (EMT) transitions. In this review, we summarize current knowledge on EOC heterogeneity in an EMT context, outline major regulators of EMT in ovarian cancer, address controversies in EMT and EOC chemoresistance, and highlight computational modeling approaches toward understanding EMT/MET in EOC.
Collapse
Affiliation(s)
- Yuliya Klymenko
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA.
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA.
| | - Oleg Kim
- Department of Applied and Computational Mathematics and Statistics, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA.
- Department of Mathematics, University of California Riverside, Riverside, CA 92521, USA.
| | - M Sharon Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA.
| |
Collapse
|
70
|
Dewhirst MW, Ashcraft KA. Implications of Increase in Vascular Permeability in Tumors by VEGF: A Commentary on the Pioneering Work of Harold Dvorak. Cancer Res 2017; 76:3118-20. [PMID: 27251086 DOI: 10.1158/0008-5472.can-16-1292] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/04/2016] [Indexed: 11/16/2022]
Abstract
See related article by Senger et al., Cancer Res 1986;46:5629-32Visit the Cancer Research 75(th) Anniversary timeline.
Collapse
|
71
|
Advanced biomaterials and microengineering technologies to recapitulate the stepwise process of cancer metastasis. Biomaterials 2017; 133:176-207. [DOI: 10.1016/j.biomaterials.2017.04.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/04/2017] [Accepted: 04/12/2017] [Indexed: 02/08/2023]
|
72
|
Evje S. An integrative multiphase model for cancer cell migration under influence of physical cues from the microenvironment. Chem Eng Sci 2017. [DOI: 10.1016/j.ces.2017.02.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
73
|
Nerger BA, Siedlik MJ, Nelson CM. Microfabricated tissues for investigating traction forces involved in cell migration and tissue morphogenesis. Cell Mol Life Sci 2017; 74:1819-1834. [PMID: 28008471 PMCID: PMC5391279 DOI: 10.1007/s00018-016-2439-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/02/2016] [Accepted: 12/08/2016] [Indexed: 01/09/2023]
Abstract
Cell-generated forces drive an array of biological processes ranging from wound healing to tumor metastasis. Whereas experimental techniques such as traction force microscopy are capable of quantifying traction forces in multidimensional systems, the physical mechanisms by which these forces induce changes in tissue form remain to be elucidated. Understanding these mechanisms will ultimately require techniques that are capable of quantifying traction forces with high precision and accuracy in vivo or in systems that recapitulate in vivo conditions, such as microfabricated tissues and engineered substrata. To that end, here we review the fundamentals of traction forces, their quantification, and the use of microfabricated tissues designed to study these forces during cell migration and tissue morphogenesis. We emphasize the differences between traction forces in two- and three-dimensional systems, and highlight recently developed techniques for quantifying traction forces.
Collapse
Affiliation(s)
- Bryan A Nerger
- Department of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ, 08544, USA
| | - Michael J Siedlik
- Department of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ, 08544, USA
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ, 08544, USA.
- Department of Molecular Biology, Princeton University, 303 Hoyt Laboratory, William Street, Princeton, NJ, 08544, USA.
| |
Collapse
|
74
|
Xu L, Li J, Bao Z, Xu P, Chang H, Wu J, Bei Y, Xia L, Wu P, Yan K, Lu B, Cui G. Silencing of OTUB1 inhibits migration of human glioma cells in vitro. Neuropathology 2017; 37:217-226. [PMID: 28139865 DOI: 10.1111/neup.12366] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 01/12/2023]
Affiliation(s)
- Li Xu
- Department of Neurosurgery; The First Affiliated Hospital of Soochow University; Suzhou Jiangsu Province China
| | - Jinquan Li
- Department of Neurosurgery; The First Affiliated Hospital of Soochow University; Suzhou Jiangsu Province China
| | - Zhen Bao
- Department of Neurosurgery; The First Affiliated Hospital of Soochow University; Suzhou Jiangsu Province China
| | - Peng Xu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong Jiangsu Province China
| | - Hao Chang
- Department of Neurosurgery; Affiliated Wuxi Second Hospital of Nanjing Medical University; Wuxi Jiangsu Province China
| | - Jingjing Wu
- Department of Oncology; Nantong Rich Hospital; Nantong Jiangsu Province China
| | - Yuanqi Bei
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong Jiangsu Province China
| | - Liuwan Xia
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong Jiangsu Province China
| | - Peizhang Wu
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target; Nantong Jiangsu Province China
| | - Ke Yan
- Department of Neurosurgery; The First Affiliated Hospital of Soochow University; Suzhou Jiangsu Province China
| | - Bing Lu
- Department of Neurosurgery; The First Affiliated Hospital of Soochow University; Suzhou Jiangsu Province China
| | - Gang Cui
- Department of Neurosurgery; The First Affiliated Hospital of Soochow University; Suzhou Jiangsu Province China
| |
Collapse
|
75
|
Portillo-Lara R, Annabi N. Microengineered cancer-on-a-chip platforms to study the metastatic microenvironment. LAB ON A CHIP 2016; 16:4063-4081. [PMID: 27605305 DOI: 10.1039/c6lc00718j] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
More than 90% of cancer-related deaths can be attributed to the occurrence of metastatic diseases. Recent studies have highlighted the importance of the multicellular, biochemical and biophysical stimuli from the tumor microenvironment during carcinogenesis, treatment failure, and metastasis. Therefore, there is a need for experimental platforms that are able to recapitulate the complex pathophysiological features of the metastatic microenvironment. Recent advancements in biomaterials, microfluidics, and tissue engineering have led to the development of living multicellular microculture systems, which are maintained in controllable microenvironments and function with organ level complexity. The applications of these "on-chip" technologies for detection, separation, characterization and three dimensional (3D) propagation of cancer cells have been extensively reviewed in previous works. In this contribution, we focus on integrative microengineered platforms that allow the study of multiple aspects of the metastatic microenvironment, including the physicochemical cues from the tumor associated stroma, the heterocellular interactions that drive trans-endothelial migration and angiogenesis, the environmental stresses that metastatic cancer cells encounter during migration, and the physicochemical gradients that direct cell motility and invasion. We discuss the application of these systems as in vitro assays to elucidate fundamental mechanisms of cancer metastasis, as well as their use as human relevant platforms for drug screening in biomimetic microenvironments. We then conclude with our commentaries on current progress and future perspectives of microengineered systems for fundamental and translational cancer research.
Collapse
Affiliation(s)
- R Portillo-Lara
- Department of Chemical Engineering, Northeastern University, 451 Snell Engineering Building, 360 Huntington Ave, Boston, MA 02115, USA. and Centro de Biotecnología-FEMSA, Tecnológico de Monterrey, Monterrey, Mexico
| | - N Annabi
- Department of Chemical Engineering, Northeastern University, 451 Snell Engineering Building, 360 Huntington Ave, Boston, MA 02115, USA. and Biomaterials Innovation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA and Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|