1
|
Hemmati F, Akinpelu A, Nweze DC, Mistriotis P. 3D confinement alters smooth muscle cell responses to chemical and mechanical cues. APL Bioeng 2024; 8:046103. [PMID: 39464377 PMCID: PMC11512639 DOI: 10.1063/5.0225569] [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: 06/25/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024] Open
Abstract
Smooth muscle cell (SMC) phenotypic switching is a hallmark of many vascular diseases. Although prior work has established that chemical and mechanical cues contribute to SMC phenotypic switching, the impact of three-dimensional (3D) confinement on this process remains elusive. Yet, in vivo, arterial SMCs reside within confined environments. In this study, we designed a microfluidic assay to investigate the interplay between 3D confinement and different environmental stimuli in SMC function. Our results show that tightly, but not moderately, confined SMCs acquire a contractile phenotype when exposed to collagen I. Elevated compressive forces induced by hydrostatic pressure abolish this upregulation of the contractile phenotype and compromise SMC survival, particularly in tightly confined spaces. Transforming growth factor beta 1, which promotes the contractile state in moderate confinement, fails to enhance the contractility of tightly confined cells. Fibronectin and engagement of cadherin 2 suppress the contractile phenotype of SMCs regardless of the degree of confinement. In contrast, homophilic engagement of cadherin 11 upregulates SMC-specific genes and enhances contractility in both moderately and tightly confined cells. Overall, our work introduces 3D confinement as a regulator of SMC phenotypic responses to chemical and mechanical signals.
Collapse
Affiliation(s)
- Farnaz Hemmati
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | - Ayuba Akinpelu
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | - Daniel Chinedu Nweze
- Department of Chemical Engineering, Auburn University, Auburn, Alabama 36849, USA
| | | |
Collapse
|
2
|
Chitnis MS, Gao X, Marlena J, Holle AW. The mechanical journey of primordial germ cells. Am J Physiol Cell Physiol 2024; 327:C1532-C1545. [PMID: 39466178 DOI: 10.1152/ajpcell.00404.2024] [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: 06/13/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
Primordial germ cells (PGCs) are the earliest progenitors of germline cells of the gonads in animals. The tissues that arise from primordial germ cells give rise to male and female gametes and are thus responsible for transmitting genetic information to subsequent generations. The development of gonads, from single cells to fully formed organs, is of great interest to the reproductive biology community. In most higher animals, PGCs are initially specified at a site away from the gonads. They then migrate across multiple tissue microenvironments to reach a mesodermal mass of cells called the genital ridge, where they associate with somatic cells to form sex-specific reproductive organs. Their migratory behavior has been studied extensively to identify which tissues they interact with and how this might affect gonad development. A crucial point overlooked by classical studies has been the physical environment experienced by PGCs as they migrate and the mechanical challenges they might encounter along the way. It has long been understood that migrating cells can sense and adapt to physical forces around them via a variety of mechanisms, and studies have shown that these mechanical signals can guide stem cell fate. In this review, we summarize the mechanical microenvironment of migrating PGCs in different organisms. We describe how cells can adapt to this environment and how this adaptation can influence cell fate. Finally, we propose that mechanical signals play a crucial role in the normal development of the germline and shed light on this unexplored area of developmental biology.
Collapse
Affiliation(s)
- Malhar S Chitnis
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Xu Gao
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - Jennifer Marlena
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Andrew W Holle
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore
| |
Collapse
|
3
|
Wang C, Huangfu Y, Wang J, Lu X, Liu D, Zhang ZL. Microchip construction for migration assays: investigating the impact of physical confinement on cell morphology and motility during vaccinia virus infection. Anal Bioanal Chem 2024; 416:5605-5618. [PMID: 39158632 DOI: 10.1007/s00216-024-05485-5] [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: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/20/2024]
Abstract
Vaccinia virus (VACV)-induced cell migration is thought to be closely related to the rapid transmission of viral infection in the body. The limited studies are mainly based on scratch assay using traditional cell culture techniques, which inevitably ignores the influences of extracellular microenvironment. Physical confinement, inherently presenting in vivo, has proven to be a critical extern cue in modulating migration behaviors of multiple cells, while its impacts on VACV-induced cell motility remain unclear. Herein, we developed a migration assay microchip featuring confined microchannel array to investigate the effect of physical confinement on infected cell morphology and motility during VACV infection. Results showed that different from the random cell migration observed in traditional scratch assay on planar substrate, VACV-infected cells exhibited accelerated directionally persistent migration under confinement microenvironment. Moreover, single-directed elongated dominant lamella appeared to contrast distinctly with multiple protrusions stretched in random directions under unconfined condition. Additionally, the Golgi complex tended to relocate behind the nucleus confined within the microchannel axis compared to the classical reorientation pattern. These differences in characteristic subcellular architecture and organelle reorientation of migrating cells revealed cell biological mechanisms underlying altered migration behavior. Collectively, our study demonstrates that physical confinement acting as a guidance cue has profound impacts on VACV-induced migration behaviors, which provides new insight into cell migration behavior and viral rapid spread during VACV infection.
Collapse
Affiliation(s)
- Cheng Wang
- School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China.
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China.
| | - Yueyue Huangfu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
| | - Ji Wang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, Hong Kong, 999077, China
| | - Xiaofeng Lu
- School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Dong Liu
- School of Life Sciences, Co-Innovation Center of Neuroregeneration, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, 226019, China
| | - Zhi-Ling Zhang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
4
|
Liang L, Song X, Zhao H, Lim CT. Insights into the mechanobiology of cancer metastasis via microfluidic technologies. APL Bioeng 2024; 8:021506. [PMID: 38841688 PMCID: PMC11151435 DOI: 10.1063/5.0195389] [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: 01/02/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
During cancer metastasis, cancer cells will encounter various microenvironments with diverse physical characteristics. Changes in these physical characteristics such as tension, stiffness, viscosity, compression, and fluid shear can generate biomechanical cues that affect cancer cells, dynamically influencing numerous pathophysiological mechanisms. For example, a dense extracellular matrix drives cancer cells to reorganize their cytoskeleton structures, facilitating confined migration, while this dense and restricted space also acts as a physical barrier that potentially results in nuclear rupture. Identifying these pathophysiological processes and understanding their underlying mechanobiological mechanisms can aid in the development of more effective therapeutics targeted to cancer metastasis. In this review, we outline the advances of engineering microfluidic devices in vitro and their role in replicating tumor microenvironment to mimic in vivo settings. We highlight the potential cellular mechanisms that mediate their ability to adapt to different microenvironments. Meanwhile, we also discuss some important mechanical cues that still remain challenging to replicate in current microfluidic devices in future direction. While much remains to be explored about cancer mechanobiology, we believe the developments of microfluidic devices will reveal how these physical cues impact the behaviors of cancer cells. It will be crucial in the understanding of cancer metastasis, and potentially contributing to better drug development and cancer therapy.
Collapse
Affiliation(s)
- Lanfeng Liang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Xiao Song
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | | |
Collapse
|
5
|
Yang C, Yin D, Zhang H, Badea I, Yang SM, Zhang W. Cell Migration Assays and Their Application to Wound Healing Assays-A Critical Review. MICROMACHINES 2024; 15:720. [PMID: 38930690 PMCID: PMC11205366 DOI: 10.3390/mi15060720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
In recent years, cell migration assays (CMAs) have emerged as a tool to study the migration of cells along with their physiological responses under various stimuli, including both mechanical and bio-chemical properties. CMAs are a generic system in that they support various biological applications, such as wound healing assays. In this paper, we review the development of the CMA in the context of its application to wound healing assays. As such, the wound healing assay will be used to derive the requirements on CMAs. This paper will provide a comprehensive and critical review of the development of CMAs along with their application to wound healing assays. One salient feature of our methodology in this paper is the application of the so-called design thinking; namely we define the requirements of CMAs first and then take them as a benchmark for various developments of CMAs in the literature. The state-of-the-art CMAs are compared with this benchmark to derive the knowledge and technological gap with CMAs in the literature. We will also discuss future research directions for the CMA together with its application to wound healing assays.
Collapse
Affiliation(s)
- Chun Yang
- School of Mechanical Engineering, Donghua University, Shanghai 200051, China;
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| | - Di Yin
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China; (D.Y.); (H.Z.)
| | - Hongbo Zhang
- School of Mechanical and Power Engineering, East China University of Science and Technology, Shanghai 200237, China; (D.Y.); (H.Z.)
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada;
| | - Shih-Mo Yang
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, China
| | - Wenjun Zhang
- School of Mechanical Engineering, Donghua University, Shanghai 200051, China;
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, SK S7N 5A9, Canada
| |
Collapse
|
6
|
Atkins D, Rosas JM, Månsson LK, Shahverdi N, Dey SS, Pitenis AA. Survival-Associated Cellular Response Maintained in Pancreatic Ductal Adenocarcinoma (PDAC) Switched Between Soft and Stiff 3D Microgel Culture. ACS Biomater Sci Eng 2024; 10:2177-2187. [PMID: 38466617 PMCID: PMC11005012 DOI: 10.1021/acsbiomaterials.3c01079] [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: 08/03/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for about 90% of all pancreatic cancer cases. Five-year survival rates have remained below 12% since the 1970s, in part due to the difficulty in detection prior to metastasis (migration and invasion into neighboring organs and glands). Mechanical memory is a concept that has emerged over the past decade that may provide a path toward understanding how invading PDAC cells "remember" the mechanical properties of their diseased ("stiff", elastic modulus, E ≈ 10 kPa) microenvironment even while invading a healthy ("soft", E ≈ 1 kPa) microenvironment. Here, we investigated the role of mechanical priming by culturing a dilute suspension of PDAC (FG) cells within a 3D, rheologically tunable microgel platform from hydrogels with tunable mechanical properties. We conducted a suite of acute (short-term) priming studies where we cultured PDAC cells in either a soft (E ≈ 1 kPa) or stiff (E ≈ 10 kPa) environment for 6 h, then removed and placed them into a new soft or stiff 3D environment for another 18 h. Following these steps, we conducted RNA-seq analyses to quantify gene expression. Initial priming in the 3D culture showed persistent gene expression for the duration of the study, regardless of the subsequent environments (stiff or soft). Stiff 3D culture was associated with the downregulation of tumor suppressors (LATS1, BCAR3, CDKN2C), as well as the upregulation of cancer-associated genes (RAC3). Immunofluorescence staining (BCAR3, RAC3) further supported the persistence of this cellular response, with BCAR3 upregulated in soft culture and RAC3 upregulated in stiff-primed culture. Stiff-primed genes were stratified against patient data found in The Cancer Genome Atlas (TCGA). Upregulated genes in stiff-primed 3D culture were associated with decreased survival in patient data, suggesting a link between patient survival and mechanical priming.
Collapse
Affiliation(s)
- Dixon
J. Atkins
- Department
of Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Jonah M. Rosas
- Department
of Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Lisa K. Månsson
- Materials
Department, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| | - Nima Shahverdi
- Molecular,
Cellular, and Developmental Biology Department, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Siddharth S. Dey
- Department
of Chemical Engineering, University
of California Santa Barbara, Santa
Barbara, California 93106, United States
- Department
of Bioengineering, University of California
Santa Barbara, Santa Barbara, California 93106, United States
| | - Angela A. Pitenis
- Materials
Department, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| |
Collapse
|
7
|
Brückner DB, Broedersz CP. Learning dynamical models of single and collective cell migration: a review. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2024; 87:056601. [PMID: 38518358 DOI: 10.1088/1361-6633/ad36d2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 03/22/2024] [Indexed: 03/24/2024]
Abstract
Single and collective cell migration are fundamental processes critical for physiological phenomena ranging from embryonic development and immune response to wound healing and cancer metastasis. To understand cell migration from a physical perspective, a broad variety of models for the underlying physical mechanisms that govern cell motility have been developed. A key challenge in the development of such models is how to connect them to experimental observations, which often exhibit complex stochastic behaviours. In this review, we discuss recent advances in data-driven theoretical approaches that directly connect with experimental data to infer dynamical models of stochastic cell migration. Leveraging advances in nanofabrication, image analysis, and tracking technology, experimental studies now provide unprecedented large datasets on cellular dynamics. In parallel, theoretical efforts have been directed towards integrating such datasets into physical models from the single cell to the tissue scale with the aim of conceptualising the emergent behaviour of cells. We first review how this inference problem has been addressed in both freely migrating and confined cells. Next, we discuss why these dynamics typically take the form of underdamped stochastic equations of motion, and how such equations can be inferred from data. We then review applications of data-driven inference and machine learning approaches to heterogeneity in cell behaviour, subcellular degrees of freedom, and to the collective dynamics of multicellular systems. Across these applications, we emphasise how data-driven methods can be integrated with physical active matter models of migrating cells, and help reveal how underlying molecular mechanisms control cell behaviour. Together, these data-driven approaches are a promising avenue for building physical models of cell migration directly from experimental data, and for providing conceptual links between different length-scales of description.
Collapse
Affiliation(s)
- David B Brückner
- Institute of Science and Technology Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Chase P Broedersz
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Arnold Sommerfeld Center for Theoretical Physics and Center for NanoScience, Department of Physics, Ludwig-Maximilian-University Munich, Theresienstr. 37, D-80333 Munich, Germany
| |
Collapse
|
8
|
Kim D, Kim DH. Subcellular mechano-regulation of cell migration in confined extracellular microenvironment. BIOPHYSICS REVIEWS 2023; 4:041305. [PMID: 38505424 PMCID: PMC10903498 DOI: 10.1063/5.0185377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/01/2023] [Indexed: 03/21/2024]
Abstract
Cell migration is a highly coordinated cellular event that determines diverse physiological and pathological processes in which the continuous interaction of a migrating cell with neighboring cells or the extracellular matrix is regulated by the physical setting of the extracellular microenvironment. In confined spaces, cell migration occurs differently compared to unconfined open spaces owing to the additional forces that limit cell motility, which create a driving bias for cells to invade the confined space, resulting in a distinct cell motility process compared to what is expected in open spaces. Moreover, cells in confined environments can be subjected to elevated mechanical compression, which causes physical stimuli and activates the damage repair cycle in the cell, including the DNA in the nucleus. Although cells have a self-restoring system to repair damage from the cell membrane to the genetic components of the nucleus, this process may result in genetic and/or epigenetic alterations that can increase the risk of the progression of diverse diseases, such as cancer and immune disorders. Furthermore, there has been a shift in the paradigm of bioengineering from the development of new biomaterials to controlling biophysical cues and fine-tuning cell behaviors to cure damaged/diseased tissues. The external physical cues perceived by cells are transduced along the mechanosensitive machinery, which is further channeled into the nucleus through subcellular molecular linkages of the nucleoskeleton and cytoskeleton or the biochemical translocation of transcription factors. Thus, external cues can directly or indirectly regulate genetic transcriptional processes and nuclear mechanics, ultimately determining cell fate. In this review, we discuss the importance of the biophysical cues, response mechanisms, and mechanical models of cell migration in confined environments. We also discuss the effect of force-dependent deformation of subcellular components, specifically focusing on subnuclear organelles, such as nuclear membranes and chromosomal organization. This review will provide a biophysical perspective on cancer progression and metastasis as well as abnormal cellular proliferation.
Collapse
Affiliation(s)
- Daesan Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | | |
Collapse
|
9
|
Li Y, Jiang W, Zhou X, Long Y, Sun Y, Zeng Y, Yao X. Advances in Regulating Cellular Behavior Using Micropatterns. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2023; 96:527-547. [PMID: 38161579 PMCID: PMC10751872 DOI: 10.59249/uxoh1740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Micropatterns, characterized as distinct physical microstructures or chemical adhesion matrices on substance surfaces, have emerged as a powerful tool for manipulating cellular activity. By creating specific extracellular matrix microenvironments, micropatterns can influence various cell behaviors, including orientation, proliferation, migration, and differentiation. This review provides a comprehensive overview of the latest advancements in the use of micropatterns for cell behavior regulation. It discusses the influence of micropattern morphology and coating on cell behavior and the underlying mechanisms. It also highlights future research directions in this field, aiming to inspire new investigations in materials medicine, regenerative medicine, and tissue engineering. The review underscores the potential of micropatterns as a novel approach for controlling cell behavior, which could pave the way for breakthroughs in various biomedical applications.
Collapse
Affiliation(s)
- Yizhou Li
- Institute of Biomedical Engineering, West China School
of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu,
P.R. China
- State Key Laboratory of Oral Diseases & National
Center for Stomatology & National Clinical Research Center for Oral
Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, P.R.
China
| | - Wenli Jiang
- Institute of Biomedical Engineering, West China School
of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu,
P.R. China
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School
of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu,
P.R. China
| | - Yicen Long
- Institute of Biomedical Engineering, West China School
of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu,
P.R. China
| | - Yujia Sun
- Institute of Biomedical Engineering, West China School
of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu,
P.R. China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School
of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu,
P.R. China
| | - Xinghong Yao
- Radiation Oncology Key Laboratory of Sichuan Province,
Department of Radiotherapy, Sichuan Clinical Research Center for Cancer, Sichuan
Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital
of University of Electronic Science and Technology of China, Chengdu, P.R.
China
| |
Collapse
|
10
|
Banerjee S, Smith IM, Hengen AC, Stroka KM. Methods for studying mammalian aquaporin biology. Biol Methods Protoc 2023; 8:bpad031. [PMID: 38046463 PMCID: PMC10689382 DOI: 10.1093/biomethods/bpad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/29/2023] [Accepted: 11/09/2023] [Indexed: 12/05/2023] Open
Abstract
Aquaporins (AQPs), transmembrane water-conducting channels, have earned a great deal of scrutiny for their critical physiological roles in healthy and disease cell states, especially in the biomedical field. Numerous methods have been implemented to elucidate the involvement of AQP-mediated water transport and downstream signaling activation in eliciting whole cell, tissue, and organ functional responses. To modulate these responses, other methods have been employed to investigate AQP druggability. This review discusses standard in vitro, in vivo, and in silico methods for studying AQPs, especially for biomedical and mammalian cell biology applications. We also propose some new techniques and approaches for future AQP research to address current gaps in methodology.
Collapse
Affiliation(s)
- Shohini Banerjee
- Fischell Department of Bioengineering, University of Maryland, MD 20742, United States
| | - Ian M Smith
- Fischell Department of Bioengineering, University of Maryland, MD 20742, United States
| | - Autumn C Hengen
- Fischell Department of Bioengineering, University of Maryland, MD 20742, United States
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, MD 20742, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore MD 21201, United States
- Biophysics Program, University of Maryland, MD 20742, United States
- Center for Stem Cell Biology and Regenerative Medicine, University of Maryland, Baltimore MD 21201, United States
| |
Collapse
|
11
|
Martínez-Calvo A, Trenado-Yuste C, Lee H, Gore J, Wingreen NS, Datta SS. Interfacial morphodynamics of proliferating microbial communities. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563665. [PMID: 37961366 PMCID: PMC10634769 DOI: 10.1101/2023.10.23.563665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
In microbial communities, various cell types often coexist by occupying distinct spatial domains. What determines the shape of the interface between such domains-which in turn influences the interactions between cells and overall community function? Here, we address this question by developing a continuum model of a 2D spatially-structured microbial community with two distinct cell types. We find that, depending on the balance of the different cell proliferation rates and substrate friction coefficients, the interface between domains is either stable and smooth, or unstable and develops finger-like protrusions. We establish quantitative principles describing when these different interfacial behaviors arise, and find good agreement both with the results of previous experimental reports as well as new experiments performed here. Our work thus helps to provide a biophysical basis for understanding the interfacial morphodynamics of proliferating microbial communities, as well as a broader range of proliferating active systems.
Collapse
|
12
|
Hemmati F, Akinpelu A, Song J, Amiri F, McDaniel A, McMurray C, Afthinos A, Andreadis ST, Aitken AV, Biancardi VC, Gerecht S, Mistriotis P. Downregulation of YAP Activity Restricts P53 Hyperactivation to Promote Cell Survival in Confinement. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302228. [PMID: 37267923 PMCID: PMC10427377 DOI: 10.1002/advs.202302228] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Indexed: 06/04/2023]
Abstract
Cell migration through confining three dimensional (3D) topographies can lead to loss of nuclear envelope integrity, DNA damage, and genomic instability. Despite these detrimental phenomena, cells transiently exposed to confinement do not usually die. Whether this is also true for cells subjected to long-term confinement remains unclear at present. To investigate this, photopatterning and microfluidics are employed to fabricate a high-throughput device that circumvents limitations of previous cell confinement models and enables prolonged culture of single cells in microchannels with physiologically relevant length scales. The results of this study show that continuous exposure to tight confinement can trigger frequent nuclear envelope rupture events, which in turn promote P53 activation and cell apoptosis. Migrating cells eventually adapt to confinement and evade cell death by downregulating YAP activity. Reduced YAP activity, which is the consequence of confinement-induced YAP1/2 translocation to the cytoplasm, suppresses the incidence of nuclear envelope rupture and abolishes P53-mediated cell death. Cumulatively, this work establishes advanced, high-throughput biomimetic models for better understanding cell behavior in health and disease, and underscores the critical role of topographical cues and mechanotransduction pathways in the regulation of cell life and death.
Collapse
Affiliation(s)
- Farnaz Hemmati
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | - Ayuba Akinpelu
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Farshad Amiri
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | - Anya McDaniel
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | - Collins McMurray
- Department of Chemical EngineeringAuburn UniversityAuburnAL36849USA
| | | | - Stelios T. Andreadis
- Departments of Chemical and Biological EngineeringThe State University of New YorkBuffaloNY14260USA
- Department of Biomedical EngineeringUniversity at BuffaloThe State University of New YorkBuffaloNY14228USA
- Center of Excellence in Bioinformatics and Life SciencesBuffaloNY14203USA
- Center for Cell Gene and Tissue Engineering (CGTE)University at BuffaloThe State University of New YorkBuffaloNY14260USA
| | - Andrew V. Aitken
- Department of AnatomyPhysiology and PharmacologyCollege of Veterinary MedicineAuburn UniversityAuburnAL36849USA
- Center for Neurosciences InitiativeAuburn UniversityAuburnAL36849USA
| | - Vinicia C. Biancardi
- Department of AnatomyPhysiology and PharmacologyCollege of Veterinary MedicineAuburn UniversityAuburnAL36849USA
- Center for Neurosciences InitiativeAuburn UniversityAuburnAL36849USA
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | |
Collapse
|
13
|
Hu Y, Becker ML, Willits RK. Quantification of cell migration: metrics selection to model application. Front Cell Dev Biol 2023; 11:1155882. [PMID: 37255596 PMCID: PMC10225508 DOI: 10.3389/fcell.2023.1155882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/05/2023] [Indexed: 06/01/2023] Open
Abstract
Cell migration plays an essential role in physiological and pathological states, such as immune response, tissue generation and tumor development. This phenomenon can occur spontaneously or it can be triggered by an external stimuli, including biochemical, mechanical, or electrical cues that induce or direct cells to migrate. The migratory response to these cues is foundational to several fields including neuroscience, cancer and regenerative medicine. Various platforms are available to qualitatively and quantitatively measure cell migration, making the measurements of cell motility straight-forward. Migratory behavior must be analyzed by multiple metrics and then models to connect the measurements to physiological meaning. This review will focus on describing and quantifying cell movement for individual cell migration.
Collapse
Affiliation(s)
- Yang Hu
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, United States
| | - Matthew L. Becker
- Departments of Chemistry, Mechanical Engineering and Materials Science, Biomedical Engineering and Orthopedic Surgery, Duke University, Durham, NC, United States
| | - Rebecca Kuntz Willits
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, United States
- Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, United States
| |
Collapse
|
14
|
Yang Z, Zhou Z, Si T, Zhou Z, Zhou L, Chin YR, Zhang L, Guan X, Yang M. High Throughput Confined Migration Microfluidic Device for Drug Screening. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2207194. [PMID: 36634971 DOI: 10.1002/smll.202207194] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/22/2022] [Indexed: 06/17/2023]
Abstract
Cancer metastasis is the major cause of cancer-related death. Excessive extracellular matrix deposition and increased stiffness are typical features of solid tumors, creating confined spaces for tumor cell migration and metastasis. Confined migration is involved in all metastasis steps. However, confined and unconfined migration inhibitors are different and drugs available to inhibit confined migration are rare. The main challenges are the modeling of confined migration, the suffering of low throughput, and others. Microfluidic device has the advantage to reduce reagent consumption and enhance throughput. Here, a microfluidic chip that can achieve multi-function drug screening against the collective migration of cancer cells under confined environment is designed. This device is applied to screen out effective drugs on confined migration among a novel mechanoreceptors compound library (166 compounds) in hepatocellular carcinoma, non-small lung cancer, breast cancer, and pancreatic ductal adenocarcinoma cells. Three compounds that can significantly inhibit confined migration in pan-cancer: mitochonic acid 5 (MA-5), SB-705498, and diphenyleneiodonium chloride are found. Finally, it is elucidated that these drugs targeted mitochondria, actin polymerization, and cell viability, respectively. In sum, a high-throughput microfluidic platform for screening drugs targeting confined migration is established and three novel inhibitors of confined migration in multiple cancer types are identified.
Collapse
Affiliation(s)
- Zihan Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, 518000, P. R. China
| | - Zhihang Zhou
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Tongxu Si
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, 518000, P. R. China
| | - Zhengdong Zhou
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, 518000, P. R. China
| | - Li Zhou
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
- Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Y Rebecca Chin
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
| | - Liang Zhang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
| | - Xinyuan Guan
- Department of Clinical Oncology, the University of Hong Kong, Hong Kong, Hong Kong SAR, 999077, P. R. China
| | - Mengsu Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, 999077, P. R. China
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong, 518000, P. R. China
| |
Collapse
|
15
|
Mukherjee A, Ron JE, Hu HT, Nishimura T, Hanawa‐Suetsugu K, Behkam B, Mimori‐Kiyosue Y, Gov NS, Suetsugu S, Nain AS. Actin Filaments Couple the Protrusive Tips to the Nucleus through the I-BAR Domain Protein IRSp53 during the Migration of Cells on 1D Fibers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207368. [PMID: 36698307 PMCID: PMC9982589 DOI: 10.1002/advs.202207368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Indexed: 05/31/2023]
Abstract
The cell migration cycle, well-established in 2D, proceeds with forming new protrusive structures at the cell membrane and subsequent redistribution of contractile machinery. Three-dimensional (3D) environments are complex and composed of 1D fibers, and 1D fibers are shown to recapitulate essential features of 3D migration. However, the establishment of protrusive activity at the cell membrane and contractility in 1D fibrous environments remains partially understood. Here the role of membrane curvature regulator IRSp53 is examined as a coupler between actin filaments and plasma membrane during cell migration on single, suspended 1D fibers. IRSp53 depletion reduced cell-length spanning actin stress fibers that originate from the cell periphery, protrusive activity, and contractility, leading to uncoupling of the nucleus from cellular movements. A theoretical model capable of predicting the observed transition of IRSp53-depleted cells from rapid stick-slip migration to smooth and slower migration due to reduced actin polymerization at the cell edges is developed, which is verified by direct measurements of retrograde actin flow using speckle microscopy. Overall, it is found that IRSp53 mediates actin recruitment at the cellular tips leading to the establishment of cell-length spanning fibers, thus demonstrating a unique role of IRSp53 in controlling cell migration in 3D.
Collapse
Affiliation(s)
- Apratim Mukherjee
- Department of Mechanical EngineeringVirginia TechBlacksburgVA24061USA
| | - Jonathan Emanuel Ron
- Department of Chemical and Biological PhysicsWeizmann Institute of ScienceRehovot7610001Israel
| | - Hooi Ting Hu
- Division of Biological ScienceGraduate School of Science and TechnologyNara Institute of Science and TechnologyIkoma630‐0192Japan
| | - Tamako Nishimura
- Division of Biological ScienceGraduate School of Science and TechnologyNara Institute of Science and TechnologyIkoma630‐0192Japan
| | | | - Bahareh Behkam
- Department of Mechanical EngineeringVirginia TechBlacksburgVA24061USA
| | - Yuko Mimori‐Kiyosue
- Laboratory for Molecular and Cellular DynamicsRIKEN Center for Biosystems Dynamics ResearchMinatojima‐minaminachiChuo‐kuKobeHyogo650‐0047Japan
| | - Nir Shachna Gov
- Department of Chemical and Biological PhysicsWeizmann Institute of ScienceRehovot7610001Israel
| | - Shiro Suetsugu
- Division of Biological ScienceGraduate School of Science and TechnologyNara Institute of Science and TechnologyIkoma630‐0192Japan
- Data Science CenterNara Institute of Science and TechnologyIkoma630‐0192Japan
- Center for Digital Green‐innovationNara Institute of Science and TechnologyIkoma630‐0192Japan
| | | |
Collapse
|
16
|
Confined environments induce polarized paraspeckle condensates. Commun Biol 2023; 6:145. [PMID: 36737664 PMCID: PMC9898560 DOI: 10.1038/s42003-023-04528-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Cancer cells experience confinement as they navigate the tumour microenvironment during metastasis. Recent studies have revealed that the nucleus can function as a 'ruler' for measuring physical confinement via membrane tension, allowing for compression-sensitive changes in migration. Cell nuclei contain many nuclear bodies that form when their components phase separate and condense within permissive local regions within the nucleus. However, how sub-nuclear organisation and phase separation changes with cell confinement and compression is largely unknown. Here we focus on paraspeckles, stress-responsive subnuclear bodies that form by phase separation around the long non-coding RNA NEAT1. As cells entered moderate confinement, a significant increase in paraspeckle number and size was observed compared to unconfined cells. Paraspeckle polarization bias towards the leading edge was also observed in confinement, correlating with regions of euchromatin. Increasing paraspeckle abundance resulted in increases in confined migration likelihood, speed, and directionality, as well as an enhancement of paraspeckle polarization towards the leading edge. This polarization of paraspeckle condensates may play a key role in regulating confined migration and invasion in cancer cells, and illustrates the utility of microchannel-based assays for identifying phenomena not observed on 2D or 3D bulk substrates.
Collapse
|
17
|
Yang Z, Zhou L, Si T, Chen S, Liu C, Ng KK, Wang Z, Chen Z, Qiu C, Liu G, Wang Q, Zhou X, Zhang L, Yao Z, He S, Yang M, Zhou Z. Lysyl hydroxylase LH1 promotes confined migration and metastasis of cancer cells by stabilizing Septin2 to enhance actin network. Mol Cancer 2023; 22:21. [PMID: 36721170 PMCID: PMC9887875 DOI: 10.1186/s12943-023-01727-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/22/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Excessive extracellular matrix deposition and increased stiffness are typical features of solid tumors such as hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC). These conditions create confined spaces for tumor cell migration and metastasis. The regulatory mechanism of confined migration remains unclear. METHODS LC-MS was applied to determine the differentially expressed proteins between HCC tissues and corresponding adjacent tissue. Collective migration and single cell migration microfluidic devices with 6 μm-high confined channels were designed and fabricated to mimic the in vivo confined space. 3D invasion assay was created by Matrigel and Collagen I mixture treat to adherent cells. 3D spheroid formation under various stiffness environment was developed by different substitution percentage GelMA. Immunoprecipitation was performed to pull down the LH1-binding proteins, which were identified by LC-MS. Immunofluorescent staining, FRET, RT-PCR, Western blotting, FRAP, CCK-8, transwell cell migration, wound healing, orthotopic liver injection mouse model and in vivo imaging were used to evaluate the target expression and cellular phenotype. RESULTS Lysyl hydroxylase 1 (LH1) promoted the confined migration of cancer cells at both collective and single cell levels. In addition, LH1 enhanced cell invasion in a 3D biomimetic model and spheroid formation in stiffer environments. High LH1 expression correlated with poor prognosis of both HCC and PDAC patients, while it also promoted in vivo metastasis. Mechanistically, LH1 bound and stabilized Septin2 (SEPT2) to enhance actin polymerization, depending on the hydroxylase domain. Finally, the subpopulation with high expression of both LH1 and SEPT2 had the poorest prognosis. CONCLUSIONS LH1 promotes the confined migration and metastasis of cancer cells by stabilizing SEPT2 and thus facilitating actin polymerization.
Collapse
Affiliation(s)
- Zihan Yang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Li Zhou
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tongxu Si
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Siyuan Chen
- grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chengxi Liu
- grid.16890.360000 0004 1764 6123State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Kelvin Kaki Ng
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Zesheng Wang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Zhiji Chen
- grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chan Qiu
- grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guopan Liu
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Qingliang Wang
- grid.412461.40000 0004 9334 6536Department of Pathology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyu Zhou
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Liang Zhang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Zhongping Yao
- grid.16890.360000 0004 1764 6123State Key Laboratory of Chemical Biology and Drug Discovery, Research Institute for Future Food and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Song He
- grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengsu Yang
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.35030.350000 0004 1792 6846Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Futian Research Institute, Shenzhen, Guangdong China
| | - Zhihang Zhou
- grid.35030.350000 0004 1792 6846Department of Biomedical Sciences, and Tung Biomedical Sciences Center, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, People’s Republic of China ,grid.412461.40000 0004 9334 6536Department of Gastroenterology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
18
|
Margadant C. Cell Migration in Three Dimensions. Methods Mol Biol 2023; 2608:1-14. [PMID: 36653698 DOI: 10.1007/978-1-0716-2887-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cell migration plays an essential role in many pathophysiological processes, including embryonic development, wound healing, immunity, and cancer invasion, and is therefore a widely studied phenomenon in many different fields from basic cell biology to regenerative medicine. During the past decades, a multitude of increasingly complex methods have been developed to study cell migration. Here we compile a series of current state-of-the-art methods and protocols to investigate cell migration in a variety of model systems ranging from cells, organoids, tissue explants, and microfluidic systems to Drosophila, zebrafish, and mice. Together they cover processes as diverse as nuclear deformation, energy consumption, endocytic trafficking, and matrix degradation, as well as tumor vascularization and cancer cell invasion, sprouting angiogenesis, and leukocyte extravasation. Furthermore, methods to study developmental processes such as neural tube closure, germ layer specification, and branching morphogenesis are included, as well as scripts for the automated analysis of several aspects of cell migration. Together, this book constitutes a unique collection of methods of prime importance to those interested in the analysis of cell migration.
Collapse
Affiliation(s)
- Coert Margadant
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Li Z, Jiang Z, Lu L, Liu Y. Microfluidic Manipulation for Biomedical Applications in the Central and Peripheral Nervous Systems. Pharmaceutics 2023; 15:pharmaceutics15010210. [PMID: 36678839 PMCID: PMC9862045 DOI: 10.3390/pharmaceutics15010210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/04/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Physical injuries and neurodegenerative diseases often lead to irreversible damage to the organizational structure of the central nervous system (CNS) and peripheral nervous system (PNS), culminating in physiological malfunctions. Investigating these complex and diverse biological processes at the macro and micro levels will help to identify the cellular and molecular mechanisms associated with nerve degeneration and regeneration, thereby providing new options for the development of new therapeutic strategies for the functional recovery of the nervous system. Due to their distinct advantages, modern microfluidic platforms have significant potential for high-throughput cell and organoid cultures in vitro, the synthesis of a variety of tissue engineering scaffolds and drug carriers, and observing the delivery of drugs at the desired speed to the desired location in real time. In this review, we first introduce the types of nerve damage and the repair mechanisms of the CNS and PNS; then, we summarize the development of microfluidic platforms and their application in drug carriers. We also describe a variety of damage models, tissue engineering scaffolds, and drug carriers for nerve injury repair based on the application of microfluidic platforms. Finally, we discuss remaining challenges and future perspectives with regard to the promotion of nerve injury repair based on engineered microfluidic platform technology.
Collapse
|
20
|
Abstract
Immune responses are governed by signals from the tissue microenvironment, and in addition to biochemical signals, mechanical cues and forces arising from the tissue, its extracellular matrix and its constituent cells shape immune cell function. Indeed, changes in biophysical properties of tissue alter the mechanical signals experienced by cells in many disease conditions, in inflammatory states and in the context of ageing. These mechanical cues are converted into biochemical signals through the process of mechanotransduction, and multiple pathways of mechanotransduction have been identified in immune cells. Such pathways impact important cellular functions including cell activation, cytokine production, metabolism, proliferation and trafficking. Changes in tissue mechanics may also represent a new form of 'danger signal' that alerts the innate and adaptive immune systems to the possibility of injury or infection. Tissue mechanics can change temporally during an infection or inflammatory response, offering a novel layer of dynamic immune regulation. Here, we review the emerging field of mechanoimmunology, focusing on how mechanical cues at the scale of the tissue environment regulate immune cell behaviours to initiate, propagate and resolve the immune response.
Collapse
|
21
|
Castillo Ransanz L, Van Altena PFJ, Heine VM, Accardo A. Engineered cell culture microenvironments for mechanobiology studies of brain neural cells. Front Bioeng Biotechnol 2022; 10:1096054. [PMID: 36588937 PMCID: PMC9794772 DOI: 10.3389/fbioe.2022.1096054] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/29/2022] [Indexed: 12/15/2022] Open
Abstract
The biomechanical properties of the brain microenvironment, which is composed of different neural cell types, the extracellular matrix, and blood vessels, are critical for normal brain development and neural functioning. Stiffness, viscoelasticity and spatial organization of brain tissue modulate proliferation, migration, differentiation, and cell function. However, the mechanical aspects of the neural microenvironment are largely ignored in current cell culture systems. Considering the high promises of human induced pluripotent stem cell- (iPSC-) based models for disease modelling and new treatment development, and in light of the physiological relevance of neuromechanobiological features, applications of in vitro engineered neuronal microenvironments should be explored thoroughly to develop more representative in vitro brain models. In this context, recently developed biomaterials in combination with micro- and nanofabrication techniques 1) allow investigating how mechanical properties affect neural cell development and functioning; 2) enable optimal cell microenvironment engineering strategies to advance neural cell models; and 3) provide a quantitative tool to assess changes in the neuromechanobiological properties of the brain microenvironment induced by pathology. In this review, we discuss the biological and engineering aspects involved in studying neuromechanobiology within scaffold-free and scaffold-based 2D and 3D iPSC-based brain models and approaches employing primary lineages (neural/glial), cell lines and other stem cells. Finally, we discuss future experimental directions of engineered microenvironments in neuroscience.
Collapse
Affiliation(s)
- Lucía Castillo Ransanz
- Department of Child and Adolescence Psychiatry, Amsterdam Neuroscience, Emma Children’s Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Pieter F. J. Van Altena
- Department of Precision and Microsystems Engineering, Delft University of Technology, Delft, Netherlands
| | - Vivi M. Heine
- Department of Child and Adolescence Psychiatry, Amsterdam Neuroscience, Emma Children’s Hospital, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Department of Complex Trait Genetics, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Angelo Accardo
- Department of Precision and Microsystems Engineering, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
22
|
Zarubova J, Hasani-Sadrabadi MM, Norris SCP, Majedi FS, Xiao C, Kasko AM, Li S. Cell-Taxi: Mesenchymal Cells Carry and Transport Clusters of Cancer Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203515. [PMID: 36307906 PMCID: PMC9772300 DOI: 10.1002/smll.202203515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/09/2022] [Indexed: 06/16/2023]
Abstract
Cell clusters that collectively migrate from primary tumors appear to be far more potent in forming distant metastases than single cancer cells. A better understanding of the collective cell migration phenomenon and the involvement of various cell types during this process is needed. Here, an in vitro platform based on inverted-pyramidal microwells to follow and quantify the collective migration of hundreds of tumor cell clusters at once is developed. These results indicate that mesenchymal stromal cells (MSCs) or cancer-associated fibroblasts (CAFs) in the heterotypic tumor cell clusters may facilitate metastatic dissemination by transporting low-motile cancer cells in a Rac-dependent manner and that extracellular vesicles secreted by mesenchymal cells only play a minor role in this process. Furthermore, in vivo studies show that cancer cell spheroids containing MSCs or CAFs have faster spreading rates. These findings highlight the active role of co-traveling stromal cells in the collective migration of tumor cell clusters and may help in developing better-targeted therapies.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague, 14220, Czech Republic
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Sam C P Norris
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Fatemeh Sadat Majedi
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Crystal Xiao
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Andrea M Kasko
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| | - Song Li
- Department of Bioengineering, University of California, 420 Westwood Plaza, 5121 Engineering V, Los Angeles, CA, 90095-1600, USA
| |
Collapse
|
23
|
Abstract
The morphogenesis of two-dimensional bacterial colonies has been well studied. However, little is known about the colony morphologies of bacteria growing in three dimensions, despite the prevalence of three-dimensional environments (e.g., soil, inside hosts) as natural bacterial habitats. Using experiments on bacteria in granular hydrogel matrices, we find that dense multicellular colonies growing in three dimensions undergo a common morphological instability and roughen, adopting a characteristic broccoli-like morphology when they exceed a critical size. Analysis of a continuum “active fluid” model of the expanding colony reveals that this behavior originates from an interplay of competition for nutrients with growth-driven colony expansion, both of which vary spatially. These results shed light on the fundamental biophysical principles underlying growth in three dimensions. How do growing bacterial colonies get their shapes? While colony morphogenesis is well studied in two dimensions, many bacteria grow as large colonies in three-dimensional (3D) environments, such as gels and tissues in the body or subsurface soils and sediments. Here, we describe the morphodynamics of large colonies of bacteria growing in three dimensions. Using experiments in transparent 3D granular hydrogel matrices, we show that dense colonies of four different species of bacteria generically become morphologically unstable and roughen as they consume nutrients and grow beyond a critical size—eventually adopting a characteristic branched, broccoli-like morphology independent of variations in the cell type and environmental conditions. This behavior reflects a key difference between two-dimensional (2D) and 3D colonies; while a 2D colony may access the nutrients needed for growth from the third dimension, a 3D colony inevitably becomes nutrient limited in its interior, driving a transition to unstable growth at its surface. We elucidate the onset of the instability using linear stability analysis and numerical simulations of a continuum model that treats the colony as an “active fluid” whose dynamics are driven by nutrient-dependent cellular growth. We find that when all dimensions of the colony substantially exceed the nutrient penetration length, nutrient-limited growth drives a 3D morphological instability that recapitulates essential features of the experimental observations. Our work thus provides a framework to predict and control the organization of growing colonies—as well as other forms of growing active matter, such as tumors and engineered living materials—in 3D environments.
Collapse
|
24
|
Afthinos A, Bera K, Chen J, Ozcelikkale A, Amitrano A, Choudhury MI, Huang R, Pachidis P, Mistriotis P, Chen Y, Konstantopoulos K. Migration and 3D Traction Force Measurements inside Compliant Microchannels. NANO LETTERS 2022; 22:7318-7327. [PMID: 36112517 PMCID: PMC9872269 DOI: 10.1021/acs.nanolett.2c01261] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Cells migrate in vivo through channel-like tracks. While polydimethylsiloxane devices emulate such tracks in vitro, their channel walls are impermeable and have supraphysiological stiffness. Existing hydrogel-based platforms address these issues but cannot provide high-throughput analysis of cell motility in independently controllable stiffness and confinement. We herein develop polyacrylamide (PA)-based microchannels of physiological stiffness and prescribed dimensions for high-throughput analysis of cell migration and identify a biphasic dependence of speed upon confinement and stiffness. By utilizing novel four-walled microchannels with heterogeneous stiffness, we reveal the distinct contributions of apicolateral versus basal microchannel wall stiffness to confined versus unconfined migration. While the basal wall stiffness dictates unconfined migration, apicolateral stiffness controls confined migration. By tracking nanobeads embedded within channel walls, we innovate three-dimensional traction force measurements around spatially confining cells at subcellular resolution. Our unique and highly customizable device fabrication strategy provides a physiologically relevant in vitro platform to study confined cells.
Collapse
Affiliation(s)
- Alexandros Afthinos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
| | - Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
| | - Junjie Chen
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Center for Cell Dynamics, The Johns Hopkins University, Baltimore MD, 21205, USA
| | - Altug Ozcelikkale
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Mechanical Engineering, Middle East Technical University, 06531 Ankara, Turkey
| | - Alice Amitrano
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
| | - Mohammad Ikbal Choudhury
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
| | - Randy Huang
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Center for Cell Dynamics, The Johns Hopkins University, Baltimore MD, 21205, USA
| | - Pavlos Pachidis
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Chemical Engineering, Auburn University, Auburn AL, 36849, USA
| | - Yun Chen
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Center for Cell Dynamics, The Johns Hopkins University, Baltimore MD, 21205, USA
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore MD, 21218, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore MD, 21205, USA
| |
Collapse
|
25
|
Sentoku M, Iida K, Hashimoto H, Yasuda K. Dominant geometrical factors of collective cell migration in flexible 3D gelatin tube structures. BIOPHYSICAL REPORTS 2022; 2:100063. [PMID: 36425328 PMCID: PMC9680702 DOI: 10.1016/j.bpr.2022.100063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/15/2022] [Indexed: 06/16/2023]
Abstract
Collective cell migration is a dynamic and interactive behavior of cell cohorts essential for diverse physiological developments in living organisms. Recent studies have revealed the importance of three-dimensional (3D) topographical confinements to regulate the migration modes of cell cohorts in tubular confinement. However, conventional in vitro assays fail to observe cells' behavior in response to 3D structural changes, which is necessary for examining the geometric regulation factors of collective migration. Here, we introduce a newly developed assay for fabricating flexible 3D structures of capillary microtunnels to examine the behavior of vascular endothelial cells (ECs) as they progress through the successive transition across wide or narrow tube structures. The microtunnels with altered diameters were formed inside gelatin-gel blocks by photo-thermal etching with micrometer-sized spot heating of the focused infrared laser absorption. The ECs migrated and spread two-dimensionally on the inner surface of gelatin capillary microtunnels as a monolayer instead of filling the entire capillary. In the straight cylindrical topographical constraint, leading ECs exhibited no apparent diameter dependence for the maximum peak migration velocity. However, widening the diameter in the narrow-wide structures caused a decrease in migration velocity following in direct proportion to the diameter increase ratio, whereas narrowing the diameter in wide-narrow microtunnels increased the speed without obvious correlation between velocity change and diameter change. The results demonstrated the ability of the newly developed flexible 3D gelatin tube structures for collective cell migration, and the findings provide insights into the dominant geometric factor of the emerging migratory modes for endothelial migration as asymmetric fluid flow-like behavior in the borderless cylindrical cell sheets.
Collapse
Affiliation(s)
- Mitsuru Sentoku
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku, Tokyo, Japan
| | - Kento Iida
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku, Tokyo, Japan
| | - Hiromichi Hashimoto
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku, Tokyo, Japan
| | - Kenji Yasuda
- Department of Pure and Applied Physics, Graduate School of Advanced Science and Engineering, Waseda University, Shinjuku, Tokyo, Japan
- Department of Physics, School of Advanced Science and Engineering, Waseda University, Shinjuku, Tokyo, Japan
| |
Collapse
|
26
|
Wang D, Gust M, Ferrell N. Kidney-on-a-Chip: Mechanical Stimulation and Sensor Integration. SENSORS (BASEL, SWITZERLAND) 2022; 22:6889. [PMID: 36146238 PMCID: PMC9503911 DOI: 10.3390/s22186889] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 06/16/2023]
Abstract
Bioengineered in vitro models of the kidney offer unprecedented opportunities to better mimic the in vivo microenvironment. Kidney-on-a-chip technology reproduces 2D or 3D features which can replicate features of the tissue architecture, composition, and dynamic mechanical forces experienced by cells in vivo. Kidney cells are exposed to mechanical stimuli such as substrate stiffness, shear stress, compression, and stretch, which regulate multiple cellular functions. Incorporating mechanical stimuli in kidney-on-a-chip is critically important for recapitulating the physiological or pathological microenvironment. This review will explore approaches to applying mechanical stimuli to different cell types using kidney-on-a-chip models and how these systems are used to study kidney physiology, model disease, and screen for drug toxicity. We further discuss sensor integration into kidney-on-a-chip for monitoring cellular responses to mechanical or other pathological stimuli. We discuss the advantages, limitations, and challenges associated with incorporating mechanical stimuli in kidney-on-a-chip models for a variety of applications. Overall, this review aims to highlight the importance of mechanical stimuli and sensor integration in the design and implementation of kidney-on-a-chip devices.
Collapse
Affiliation(s)
- Dan Wang
- Division of Nephrology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Matthew Gust
- Division of Nephrology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Statistics, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Nicholas Ferrell
- Division of Nephrology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
27
|
Li Y, Wong IY, Guo M. Reciprocity of Cell Mechanics with Extracellular Stimuli: Emerging Opportunities for Translational Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107305. [PMID: 35319155 PMCID: PMC9463119 DOI: 10.1002/smll.202107305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/20/2022] [Indexed: 06/14/2023]
Abstract
Human cells encounter dynamic mechanical cues in healthy and diseased tissues, which regulate their molecular and biophysical phenotype, including intracellular mechanics as well as force generation. Recent developments in bio/nanomaterials and microfluidics permit exquisitely sensitive measurements of cell mechanics, as well as spatiotemporal control over external mechanical stimuli to regulate cell behavior. In this review, the mechanobiology of cells interacting bidirectionally with their surrounding microenvironment, and the potential relevance for translational medicine are considered. Key fundamental concepts underlying the mechanics of living cells as well as the extracelluar matrix are first introduced. Then the authors consider case studies based on 1) microfluidic measurements of nonadherent cell deformability, 2) cell migration on micro/nano-topographies, 3) traction measurements of cells in three-dimensional (3D) matrix, 4) mechanical programming of organoid morphogenesis, as well as 5) active mechanical stimuli for potential therapeutics. These examples highlight the promise of disease diagnosis using mechanical measurements, a systems-level understanding linking molecular with biophysical phenotype, as well as therapies based on mechanical perturbations. This review concludes with a critical discussion of these emerging technologies and future directions at the interface of engineering, biology, and medicine.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, Hubei, 430074, China
| | - Ian Y Wong
- School of Engineering, Center for Biomedical Engineering, Joint Program in Cancer Biology, Brown University, 184 Hope St Box D, Providence, RI, 02912, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
28
|
Mehta P, Rahman Z, Ten Dijke P, Boukany PE. Microfluidics meets 3D cancer cell migration. Trends Cancer 2022; 8:683-697. [PMID: 35568647 DOI: 10.1016/j.trecan.2022.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 01/12/2023]
Abstract
An early step of metastasis requires a complex and coordinated migration of invasive tumor cells into the surrounding tumor microenvironment (TME), which contains extracellular matrix (ECM). It is being appreciated that 3D matrix-based microfluidic models have an advantage over conventional in vitro and animal models to study tumor progression events. Recent microfluidic models have enabled recapitulation of key mechanobiological features present within the TME to investigate collective cancer cell migration and invasion. Microfluidics also allows for functional interrogation and therapeutic manipulation of specific steps to study the dynamic aspects of tumor progression. In this review, we focus on recent developments in cancer cell migration and how microfluidic strategies have evolved to address the physiological complexities of the TME to visualize migration modes adapted by various tumor cells.
Collapse
Affiliation(s)
- Pranav Mehta
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands; Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| |
Collapse
|
29
|
Bai H, Yang J, Meng S, Liu C. Oral Microbiota-Driven Cell Migration in Carcinogenesis and Metastasis. Front Cell Infect Microbiol 2022; 12:864479. [PMID: 35573798 PMCID: PMC9103474 DOI: 10.3389/fcimb.2022.864479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/04/2022] [Indexed: 12/20/2022] Open
Abstract
The oral cavity harbors approximately 1,000 microbial species, and both pathogenic and commensal strains are involved in the development of carcinogenesis by stimulating chronic inflammation, affecting cell proliferation, and inhibiting cell apoptosis. Moreover, some substances produced by oral bacteria can also act in a carcinogenic manner. The link between oral microbiota and chronic inflammation as well as cell proliferation has been well established. Recently, increasing evidence has indicated the association of the oral microbiota with cell migration, which is crucial in regulating devastating diseases such as cancer. For instance, increased cell migration induced the spread of highly malignant cancer cells. Due to advanced technologies, the mechanistic understanding of cell migration in carcinogenesis and cancer metastasis is undergoing rapid progress. Thus, this review addressed the complexities of cell migration in carcinogenesis and cancer metastasis. We also integrate recent findings on the molecular mechanisms by which the oral microbiota regulates cell migration, with emphasis on the effect of the oral microbiota on adhesion, polarization, and guidance. Finally, we also highlight critical techniques, such as intravital microscopy and superresolution microscopy, for studies in this field.
Collapse
Affiliation(s)
- Huimin Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shu Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Shu Meng, ; Chengcheng Liu,
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Shu Meng, ; Chengcheng Liu,
| |
Collapse
|
30
|
Beeghly GF, Amofa KY, Fischbach C, Kumar S. Regulation of Tumor Invasion by the Physical Microenvironment: Lessons from Breast and Brain Cancer. Annu Rev Biomed Eng 2022; 24:29-59. [PMID: 35119915 DOI: 10.1146/annurev-bioeng-110220-115419] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The success of anticancer therapies is often limited by heterogeneity within and between tumors. While much attention has been devoted to understanding the intrinsic molecular diversity of tumor cells, the surrounding tissue microenvironment is also highly complex and coevolves with tumor cells to drive clinical outcomes. Here, we propose that diverse types of solid tumors share common physical motifs that change in time and space, serving as universal regulators of malignancy. We use breast cancer and glioblastoma as instructive examples and highlight how invasion in both diseases is driven by the appropriation of structural guidance cues, contact-dependent heterotypic interactions with stromal cells, and elevated interstitial fluid pressure and flow. We discuss how engineering strategies show increasing value for measuring and modeling these physical properties for mechanistic studies. Moreover, engineered systems offer great promise for developing and testing novel therapies that improve patient prognosis by normalizing the physical tumor microenvironment. Expected final online publication date for the Annual Review of Biomedical Engineering, Volume 24 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Garrett F Beeghly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA;
| | - Kwasi Y Amofa
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA; .,Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, New York, USA
| | - Sanjay Kumar
- University of California, Berkeley-University of California, San Francisco Graduate Program in Bioengineering, Berkeley, California, USA; .,Department of Bioengineering, University of California, Berkeley, Berkeley, California, USA.,Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, California, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
31
|
Moriarty RA, Mili S, Stroka KM. RNA localization in confined cells depends on cellular mechanical activity and contributes to confined migration. iScience 2022; 25:103845. [PMID: 35198898 PMCID: PMC8850802 DOI: 10.1016/j.isci.2022.103845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/30/2021] [Accepted: 01/27/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer cells experience mechanical confining forces during metastasis and, consequently, can alter their migratory mechanisms. Localization of numerous mRNAs to cell protrusions contributes to cell polarization and migration and is controlled by proteins that can bind RNA and/or cytoskeletal elements, such as the adenomatous polyposis coli (APC). Here, we demonstrate that peripheral localization of APC-dependent RNAs in cells within confined microchannels is cell type dependent. This varying phenotype is determined by the levels of a detyrosinated tubulin network. We show that this network is regulated by mechanoactivity and that cells with mechanosensitive ion channels and increased myosin II activity direct peripheral localization of the RAB13 APC-dependent RNA. Through specific mislocalization of the RAB13 RNA, we show that peripheral RNA localization contributes to confined cell migration. Our results indicate that a cell’s mechanical activity determines its ability to peripherally target RNAs and utilize them for movement in confinement. Peripheral localization of APC-dependent RNAs in confinement depends on cell type RNA localization in confined cells is controlled by the mechanoactivity of cells RNA localization phenotype is influenced by the detyrosinated tubulin network Peripheral RNA accumulation functionally contributes to confined cell migration
Collapse
Affiliation(s)
- Rebecca A. Moriarty
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Fischell Department of Bioengineering, University of Maryland College Park, College Park, MD 20742, USA
| | - Stavroula Mili
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Corresponding author
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland College Park, College Park, MD 20742, USA
- Maryland Biophysics Program, University of Maryland College Park, College Park, MD 20742, USA
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland Baltimore, Baltimore, MD 21202, USA
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Baltimore, Baltimore, MD 21202, USA
- Corresponding author
| |
Collapse
|
32
|
E-Cadherin Regulates Mitochondrial Membrane Potential in Cancer Cells. Cancers (Basel) 2021; 13:cancers13205054. [PMID: 34680202 PMCID: PMC8534231 DOI: 10.3390/cancers13205054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/19/2022] Open
Abstract
Epithelial cancer cells often have unusually higher mitochondrial membrane potential (ΔΨm) than their normal counterparts, which has been associated with increased invasiveness in vitro and higher metastatic potential in vivo. However, the mechanisms by which ΔΨm in cancer cells is regulated in tumor microenvironment (TME) remain unclear. In this study, we used an in vitro micropatterning platform to recapitulate biophysical confinement cues in the TME and investigated the mechanisms by which these regulate cancer cell ΔΨm. We found that micropatterning resulted in a spatial distribution of ΔΨm, which correlated with the level of E-cadherin mediated intercellular adhesion. There was a stark contrast in the spatial distribution of ΔΨm in the micropattern of E-cadherin-negative breast cancer cells (MDA-MB-231) compared to that of the high E-cadherin expressing (MCF-7) cancer cells. Disruption and knockout of E-cadherin adhesions rescued the low ΔΨm found at the center of MCF-7 micropatterns with high E-cadherin expression, while E-cadherin overexpression in MDA-MB-231 and MCF-7 cells lowered their ΔΨm at the micropattern center. These results show that E-cadherin plays an important role in regulating the ΔΨm of cancer cells in the context of biophysical cues in TME.
Collapse
|
33
|
Hobson CM, Aaron JS, Heddleston JM, Chew TL. Visualizing the Invisible: Advanced Optical Microscopy as a Tool to Measure Biomechanical Forces. Front Cell Dev Biol 2021; 9:706126. [PMID: 34552926 PMCID: PMC8450411 DOI: 10.3389/fcell.2021.706126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 08/09/2021] [Indexed: 01/28/2023] Open
Abstract
The importance of mechanical force in biology is evident across diverse length scales, ranging from tissue morphogenesis during embryo development to mechanotransduction across single adhesion proteins at the cell surface. Consequently, many force measurement techniques rely on optical microscopy to measure forces being applied by cells on their environment, to visualize specimen deformations due to external forces, or even to directly apply a physical perturbation to the sample via photoablation or optogenetic tools. Recent developments in advanced microscopy offer improved approaches to enhance spatiotemporal resolution, imaging depth, and sample viability. These advances can be coupled with already existing force measurement methods to improve sensitivity, duration and speed, amongst other parameters. However, gaining access to advanced microscopy instrumentation and the expertise necessary to extract meaningful insights from these techniques is an unavoidable hurdle. In this Live Cell Imaging special issue Review, we survey common microscopy-based force measurement techniques and examine how they can be bolstered by emerging microscopy methods. We further explore challenges related to the accompanying data analysis in biomechanical studies and discuss the various resources available to tackle the global issue of technology dissemination, an important avenue for biologists to gain access to pre-commercial instruments that can be leveraged for biomechanical studies.
Collapse
Affiliation(s)
- Chad M. Hobson
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Jesse S. Aaron
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - John M. Heddleston
- Cleveland Clinic Florida Research and Innovation Center, Port St. Lucie, FL, United States
| | - Teng-Leong Chew
- Advanced Imaging Center, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| |
Collapse
|
34
|
Organ-Chip Models: Opportunities for Precision Medicine in Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13174487. [PMID: 34503294 PMCID: PMC8430573 DOI: 10.3390/cancers13174487] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Among all types of cancer, Pancreatic Ductal Adenocarcinoma (PDAC) has one of the lowest survival rates, partly due to the failure of current chemotherapeutics. This treatment failure can be attributed to the complicated nature of the tumor microenvironment, where the rich fibro-inflammatory responses can hinder drug delivery and efficacy at the tumor site. Moreover, the high molecular variations in PDAC create a large heterogeneity in the tumor microenvironment among patients. Current in vivo and in vitro options for drug testing are mostly ineffective in recapitulating the complex cellular interactions and individual variations in the PDAC tumor microenvironment, and as a result, they fail to provide appropriate models for individualized drug screening. Organ-on-a-chip technology combined with patient-derived organoids may provide the opportunity for developing personalized treatment options in PDAC. Abstract Pancreatic Ductal Adenocarcinoma (PDAC) is an expeditiously fatal malignancy with a five-year survival rate of 6–8%. Conventional chemotherapeutics fail in many cases due to inadequate primary response and rapidly developing resistance. This treatment failure is particularly challenging in pancreatic cancer because of the high molecular heterogeneity across tumors. Additionally, a rich fibro-inflammatory component within the tumor microenvironment (TME) limits the delivery and effectiveness of anticancer drugs, further contributing to the lack of response or developing resistance to conventional approaches in this cancer. As a result, there is an urgent need to model pancreatic cancer ex vivo to discover effective drug regimens, including those targeting the components of the TME on an individualized basis. Patient-derived three-dimensional (3D) organoid technology has provided a unique opportunity to study patient-specific cancerous epithelium. Patient-derived organoids cultured with the TME components can more accurately reflect the in vivo tumor environment. Here we present the advances in organoid technology and multicellular platforms that could allow for the development of “organ-on-a-chip” approaches to recapitulate the complex cellular interactions in PDAC tumors. We highlight the current advances of the organ-on-a-chip-based cancer models and discuss their potential for the preclinical selection of individualized treatment in PDAC.
Collapse
|
35
|
Gupta S, Patteson AE, Schwarz JM. The role of vimentin-nuclear interactions in persistent cell motility through confined spaces. NEW JOURNAL OF PHYSICS 2021; 23:093042. [PMID: 35530563 PMCID: PMC9075336 DOI: 10.1088/1367-2630/ac2550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
The ability of cells to move through small spaces depends on the mechanical properties of the cellular cytoskeleton and on nuclear deformability. In mammalian cells, the cytoskeleton is composed of three interacting, semi-flexible polymer networks: actin, microtubules, and intermediate filaments (IF). Recent experiments of mouse embryonic fibroblasts with and without vimentin have shown that the IF vimentin plays a role in confined cell motility. Here, we develop a minimal model of a cell moving through a microchannel that incorporates explicit effects of actin and vimentin and implicit effects of microtubules. Specifically, the model consists of a cell with an actomyosin cortex and a deformable cell nucleus and mechanical linkages between the two. By decreasing the amount of vimentin, we find that the cell speed increases for vimentin-null cells compared to cells with vimentin. The loss of vimentin increases nuclear deformation and alters nuclear positioning in the cell. Assuming nuclear positioning is a read-out for cell polarity, we propose a new polarity mechanism which couples cell directional motion with cytoskeletal strength and nuclear positioning and captures the abnormally persistent motion of vimentin-null cells, as observed in experiments. The enhanced persistence indicates that the vimentin-null cells are more controlled by the confinement and so less autonomous, relying more heavily on external cues than their wild-type counterparts. Our modeling results present a quantitative interpretation for recent experiments and have implications for understanding the role of vimentin in the epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Sarthak Gupta
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY USA
| | - Alison E Patteson
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY USA
| | - J M Schwarz
- Physics Department and BioInspired Institute, Syracuse University, Syracuse, NY USA
- Indian Creek Farm, Ithaca, NY USA
| |
Collapse
|
36
|
Scott M, Żychaluk K, Bearon RN. A mathematical framework for modelling 3D cell motility: applications to glioblastoma cell migration. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2021; 38:333-354. [PMID: 34189581 DOI: 10.1093/imammb/dqab009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 11/14/2022]
Abstract
The collection of 3D cell tracking data from live images of micro-tissues is a recent innovation made possible due to advances in imaging techniques. As such there is increased interest in studying cell motility in 3D in vitro model systems but a lack of rigorous methodology for analysing the resulting data sets. One such instance of the use of these in vitro models is in the study of cancerous tumours. Growing multicellular tumour spheroids in vitro allows for modelling of the tumour microenvironment and the study of tumour cell behaviours, such as migration, which improves understanding of these cells and in turn could potentially improve cancer treatments. In this paper, we present a workflow for the rigorous analysis of 3D cell tracking data, based on the persistent random walk model, but adaptable to other biologically informed mathematical models. We use statistical measures to assess the fit of the model to the motility data and to estimate model parameters and provide confidence intervals for those parameters, to allow for parametrization of the model taking correlation in the data into account. We use in silico simulations to validate the workflow in 3D before testing our method on cell tracking data taken from in vitro experiments on glioblastoma tumour cells, a brain cancer with a very poor prognosis. The presented approach is intended to be accessible to both modellers and experimentalists alike in that it provides tools for uncovering features of the data set that may suggest amendments to future experiments or modelling attempts.
Collapse
Affiliation(s)
- M Scott
- Department of Mathematical Sciences, University of Liverpool, Liverpool L69 7ZL, UK
| | - K Żychaluk
- Department of Mathematical Sciences, University of Liverpool, Liverpool L69 7ZL, UK
| | - R N Bearon
- Department of Mathematical Sciences, University of Liverpool, Liverpool L69 7ZL, UK
| |
Collapse
|
37
|
Sharma VP, Williams J, Leung E, Sanders J, Eddy R, Castracane J, Oktay MH, Entenberg D, Condeelis JS. SUN-MKL1 Crosstalk Regulates Nuclear Deformation and Fast Motility of Breast Carcinoma Cells in Fibrillar ECM Microenvironment. Cells 2021; 10:1549. [PMID: 34205257 PMCID: PMC8234170 DOI: 10.3390/cells10061549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022] Open
Abstract
Aligned collagen fibers provide topography for the rapid migration of single tumor cells (streaming migration) to invade the surrounding stroma, move within tumor nests towards blood vessels to intravasate and form distant metastases. Mechanisms of tumor cell motility have been studied extensively in the 2D context, but the mechanistic understanding of rapid single tumor cell motility in the in vivo context is still lacking. Here, we show that streaming tumor cells in vivo use collagen fibers with diameters below 3 µm. Employing 1D migration assays with matching in vivo fiber dimensions, we found a dependence of tumor cell motility on 1D substrate width, with cells moving the fastest and the most persistently on the narrowest 1D fibers (700 nm-2.5 µm). Interestingly, we also observed nuclear deformation in the absence of restricting extracellular matrix pores during high speed carcinoma cell migration in 1D, similar to the nuclear deformation observed in tumor cells in vivo. Further, we found that actomyosin machinery is aligned along the 1D axis and actomyosin contractility synchronously regulates cell motility and nuclear deformation. To further investigate the link between cell speed and nuclear deformation, we focused on the Linker of Nucleoskeleton and Cytoskeleton (LINC) complex proteins and SRF-MKL1 signaling, key regulators of mechanotransduction, actomyosin contractility and actin-based cell motility. Analysis of The Cancer Genome Atlas dataset showed a dramatic decrease in the LINC complex proteins SUN1 and SUN2 in primary tumor compared to the normal tissue. Disruption of LINC complex by SUN1 + 2 KD led to multi-lobular elongated nuclei, increased tumor cell motility and concomitant increase in F-actin, without affecting Lamin proteins. Mechanistically, we found that MKL1, an effector of changes in cellular G-actin to F-actin ratio, is required for increased 1D motility seen in SUN1 + 2 KD cells. Thus, we demonstrate a previously unrecognized crosstalk between SUN proteins and MKL1 transcription factor in modulating nuclear shape and carcinoma cell motility in an in vivo relevant 1D microenvironment.
Collapse
Affiliation(s)
- Ved P. Sharma
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - James Williams
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Edison Leung
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
| | - Joe Sanders
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Robert Eddy
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
| | - James Castracane
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA; (J.W.); (J.S.); (J.C.)
| | - Maja H. Oktay
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Entenberg
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John S. Condeelis
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.L.); (R.E.); (M.H.O.); (D.E.)
- Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
38
|
Zhou Z, Cui F, Wen Q, Susan Zhou H. Effect of vimentin on cell migration in collagen-coated microchannels: A mimetic physiological confined environment. BIOMICROFLUIDICS 2021; 15:034105. [PMID: 34025897 PMCID: PMC8133791 DOI: 10.1063/5.0045197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 05/02/2021] [Indexed: 06/12/2023]
Abstract
Cancer cell migration through tissue pores and tracks into the bloodstream is a critical biological step for cancer metastasis. Although in vivo studies have shown that expression of vimentin can induce invasive cell lines, its role in cell cytoskeleton reorganization and cell motility under in vitro physical confinement remains unknown. Here, a microfluidic device with cell culture chamber and collagen-coated microchannels was developed as an in vitro model for physiological confinement environments. Using this microchannel assay, we demonstrated that the knockdown of vimentin decreases 3T3 fibroblast cell directional migration speed in confined microchannels. Additionally, as cells form dynamic membranes that define the leading edge of motile cells, different leading edge morphologies of 3T3 fibroblast and 3T3 vimentin knockdown cells were observed. The leading edge morphology change under confinement can be explained by the effect of vimentin on cytoskeletal organization and focal adhesion. The microfluidic device integrated with a time-lapse microscope provided a new approach to study the effect of vimentin on cell adhesion, migration, and invasiveness.
Collapse
Affiliation(s)
- Zhiru Zhou
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, USA
| | - Feiyun Cui
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, USA
| | - Qi Wen
- Department of Physics, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | - H. Susan Zhou
- Department of Chemical Engineering, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts 01609, USA
| |
Collapse
|
39
|
Abstract
The physical microenvironment of cells plays a fundamental role in regulating cellular behavior and cell fate, especially in the context of cancer metastasis. For example, capillary deformation can destroy arrested circulating tumor cells while the dense extracellular matrix can form a physical barrier for invading cancer cells. Understanding how metastatic cancer cells overcome the challenges brought forth by physical confinement can help in developing better therapeutics that can put a stop to this migratory stage of the metastatic cascade. Numerous in vivo and in vitro assays have been developed to recapitulate the metastatic processes and study cancer cell migration in a confining microenvironment. In this review, we summarize some of the representative techniques and the exciting new findings. We critically review the advantages, as well as challenges associated with these tools and methodologies, and provide a guide on the applications that they are most suited for. We hope future efforts that push forward our current understanding on metastasis under confinement can lead to novel and more effective diagnostic and therapeutic strategies against this dreaded disease.
Collapse
Affiliation(s)
- Kuan Jiang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Lanfeng Liang
- Mechanobiology Institute, National University of Singapore, Singapore
| | - Chwee Teck Lim
- Mechanobiology Institute, National University of Singapore, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
| |
Collapse
|
40
|
Biofabrication of aligned structures that guide cell orientation and applications in tissue engineering. Biodes Manuf 2021. [DOI: 10.1007/s42242-020-00104-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
41
|
Yeo M, Chae S, Kim G. An in vitro model using spheroids-laden nanofibrous structures for attaining high degree of myoblast alignment and differentiation. Am J Cancer Res 2021; 11:3331-3347. [PMID: 33537090 PMCID: PMC7847672 DOI: 10.7150/thno.53928] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/16/2020] [Indexed: 12/22/2022] Open
Abstract
A spheroid is an aggregation of single cells with structural and functional characteristics similar to those of 3D native tissues, and it has been utilized as one of the typical in vitro three-dimensional (3D) cell models. Scaffold-free spheroids provide outstanding reflection of tissue complexity in a 3D in vivo-like environment, but they can neither fabricate realistic macroscale 3D complex structures without avoiding necrosis nor receive direct external stimuli (i.e., stimuli from mechanical or topographical cues). Here, we propose a spheroid-laden electrospinning process to obtain in vitro model achieved using the synergistic effect of the unique bioactive components provided by the spheroids and stimulating effects provided by the aligned nanofibers. Methods: To show the functional activity of the spheroid-laden structures, we used myoblast-spheroids to obtain skeletal muscle, comprising highly aligned myotubes, utilizing an uniaxially arranged topographical cue. The spheroid-electrospinning was used to align spheroids directly by embedding them in aligned alginate nanofibers, which were controlled with various materials and processing parameters. Results: The spheroids laden in the alginate nanofibers showed high cell viability (>90%) and was compared with that of a cell-laden alginate nanofiber that was electrospun with single cells. Consequently, the spheroids laden in the aligned nanofibers showed a significantly higher degree of myotube formation and maturation. Conclusion: Results suggested that the in vitro model using electrospun spheroids could potentially be employed to understand myogenic responses for various in vitro drug tests.
Collapse
|
42
|
Hemkemeyer SA, Vollmer V, Schwarz V, Lohmann B, Honnert U, Taha M, Schnittler HJ, Bähler M. Local Myo9b RhoGAP activity regulates cell motility. J Biol Chem 2021; 296:100136. [PMID: 33268376 PMCID: PMC7949024 DOI: 10.1074/jbc.ra120.013623] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 12/22/2022] Open
Abstract
To migrate, cells assume a polarized morphology, extending forward with a leading edge with their trailing edge retracting back toward the cell body. Both cell extension and retraction critically depend on the organization and dynamics of the actin cytoskeleton, and the small, monomeric GTPases Rac and Rho are important regulators of actin. Activation of Rac induces actin polymerization and cell extension, whereas activation of Rho enhances acto-myosin II contractility and cell retraction. To coordinate migration, these processes must be carefully regulated. The myosin Myo9b, a Rho GTPase-activating protein (GAP), negatively regulates Rho activity and deletion of Myo9b in leukocytes impairs cell migration through increased Rho activity. However, it is not known whether cell motility is regulated by global or local inhibition of Rho activity by Myo9b. Here, we addressed this question by using Myo9b-deficient macrophage-like cells that expressed different recombinant Myo9b constructs. We found that Myo9b accumulates in lamellipodial extensions generated by Rac-induced actin polymerization as a function of its motor activity. Deletion of Myo9b in HL-60-derived macrophages altered cell morphology and impaired cell migration. Reintroduction of Myo9b or Myo9b motor and GAP mutants revealed that local GAP activity rescues cell morphology and migration. In summary, Rac activation leads to actin polymerization and recruitment of Myo9b, which locally inhibits Rho activity to enhance directional cell migration.
Collapse
Affiliation(s)
- Sandra A Hemkemeyer
- Institute of Molecular Cell Biology, Westfalian Wilhelms University Münster, Münster, Germany
| | - Veith Vollmer
- Institute of Molecular Cell Biology, Westfalian Wilhelms University Münster, Münster, Germany
| | - Vera Schwarz
- Institute of Molecular Cell Biology, Westfalian Wilhelms University Münster, Münster, Germany
| | - Birgit Lohmann
- Institute of Molecular Cell Biology, Westfalian Wilhelms University Münster, Münster, Germany
| | - Ulrike Honnert
- Institute of Molecular Cell Biology, Westfalian Wilhelms University Münster, Münster, Germany
| | - Muna Taha
- Institute of Anatomy & Vascular Biology, Westfalian Wilhelms University Münster, Münster, Germany
| | - Hans-Joachim Schnittler
- Institute of Anatomy & Vascular Biology, Westfalian Wilhelms University Münster, Münster, Germany
| | - Martin Bähler
- Institute of Molecular Cell Biology, Westfalian Wilhelms University Münster, Münster, Germany.
| |
Collapse
|
43
|
Butler G, Keeton SJ, Johnson LJ, Dash PR. A phenotypic switch in the dispersal strategy of breast cancer cells selected for metastatic colonization. Proc Biol Sci 2020; 287:20202523. [PMID: 33259764 DOI: 10.1098/rspb.2020.2523] [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] [Indexed: 12/12/2022] Open
Abstract
An important question in cancer evolution concerns which traits make a cell likely to successfully metastasize. Cell motility phenotypes, mediated by cell shape change, are strong candidates. We experimentally evolved breast cancer cells in vitro for metastatic capability, using selective regimes designed to simulate stages of metastasis, then quantified their motility behaviours using computer vision. All evolved lines showed changes to motility phenotypes, and we have identified a previously unknown density-dependent motility phenotype only seen in cells selected for colonization of decellularized lung tissue. These cells increase their rate of morphological change with an increase in migration speed when local cell density is high. However, when the local cell density is low, we find the opposite relationship: the rate of morphological change decreases with an increase in migration speed. Neither the ancestral population, nor cells selected for their ability to escape or invade extracellular matrix-like environments, displays this dynamic behavioural switch. Our results suggest that cells capable of distant-site colonization may be characterized by dynamic morphological phenotypes and the capacity to respond to the local social environment.
Collapse
Affiliation(s)
- George Butler
- School of Biological Sciences, University of Reading, Reading, UK
| | - Shirley J Keeton
- School of Biological Sciences, University of Reading, Reading, UK
| | - Louise J Johnson
- School of Biological Sciences, University of Reading, Reading, UK
| | - Philip R Dash
- School of Biological Sciences, University of Reading, Reading, UK
| |
Collapse
|
44
|
Patteson AE, Carroll RJ, Iwamoto DV, Janmey PA. The vimentin cytoskeleton: when polymer physics meets cell biology. Phys Biol 2020; 18:011001. [PMID: 32992303 PMCID: PMC8240483 DOI: 10.1088/1478-3975/abbcc2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The proper functions of tissues depend on the ability of cells to withstand stress and maintain shape. Central to this process is the cytoskeleton, comprised of three polymeric networks: F-actin, microtubules, and intermediate filaments (IFs). IF proteins are among the most abundant cytoskeletal proteins in cells; yet they remain some of the least understood. Their structure and function deviate from those of their cytoskeletal partners, F-actin and microtubules. IF networks show a unique combination of extensibility, flexibility and toughness that confers mechanical resilience to the cell. Vimentin is an IF protein expressed in mesenchymal cells. This review highlights exciting new results on the physical biology of vimentin intermediate filaments and their role in allowing whole cells and tissues to cope with stress.
Collapse
Affiliation(s)
- Alison E Patteson
- Physics Department, Syracuse University, Syracuse, NY 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Robert J Carroll
- Physics Department, Syracuse University, Syracuse, NY 13244, USA
- BioInspired Institute, Syracuse University, Syracuse, NY 13244, USA
| | - Daniel V Iwamoto
- Institute for Medicine and Engineering, Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul A Janmey
- Institute for Medicine and Engineering, Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
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
|
46
|
Wisniewski EO, Mistriotis P, Bera K, Law RA, Zhang J, Nikolic M, Weiger M, Parlani M, Tuntithavornwat S, Afthinos A, Zhao R, Wirtz D, Kalab P, Scarcelli G, Friedl P, Konstantopoulos K. Dorsoventral polarity directs cell responses to migration track geometries. SCIENCE ADVANCES 2020; 6:eaba6505. [PMID: 32789173 PMCID: PMC7399493 DOI: 10.1126/sciadv.aba6505] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 06/12/2020] [Indexed: 05/02/2023]
Abstract
How migrating cells differentially adapt and respond to extracellular track geometries remains unknown. Using intravital imaging, we demonstrate that invading cells exhibit dorsoventral (top-to-bottom) polarity in vivo. To investigate the impact of dorsoventral polarity on cell locomotion through different confining geometries, we fabricated microchannels of fixed cross-sectional area, albeit with distinct aspect ratios. Vertical confinement, exerted along the dorsoventral polarity axis, induces myosin II-dependent nuclear stiffening, which results in RhoA hyperactivation at the cell poles and slow bleb-based migration. In lateral confinement, directed perpendicularly to the dorsoventral polarity axis, the absence of perinuclear myosin II fails to increase nuclear stiffness. Hence, cells maintain basal RhoA activity and display faster mesenchymal migration. In summary, by integrating microfabrication, imaging techniques, and intravital microscopy, we demonstrate that dorsoventral polarity, observed in vivo and in vitro, directs cell responses in confinement by spatially tuning RhoA activity, which controls bleb-based versus mesenchymal migration.
Collapse
Affiliation(s)
- Emily O. Wisniewski
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Panagiotis Mistriotis
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA
| | - Kaustav Bera
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Robert A. Law
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jitao Zhang
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Milos Nikolic
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Maryland Biophysics Program, Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA
| | - Michael Weiger
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maria Parlani
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Soontorn Tuntithavornwat
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Alexandros Afthinos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Runchen Zhao
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Denis Wirtz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Giuliano Scarcelli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Peter Friedl
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Cancer Genomics Centre, 3584 Utrecht, Netherlands
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
- Johns Hopkins Physical Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
47
|
Doolin MT, Moriarty RA, Stroka KM. Mechanosensing of Mechanical Confinement by Mesenchymal-Like Cells. Front Physiol 2020; 11:365. [PMID: 32390868 PMCID: PMC7193100 DOI: 10.3389/fphys.2020.00365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) and tumor cells have the unique capability to migrate out of their native environment and either home or metastasize, respectively, through extremely heterogeneous environments to a distant location. Once there, they can either aid in tissue regrowth or impart an immunomodulatory effect in the case of MSCs, or form secondary tumors in the case of tumor cells. During these journeys, cells experience physically confining forces that impinge on the cell body and the nucleus, ultimately causing a multitude of cellular changes. Most drastically, confining individual MSCs within hydrogels or confining monolayers of MSCs within agarose wells can sway MSC lineage commitment, while applying a confining compressive stress to metastatic tumor cells can increase their invasiveness. In this review, we seek to understand the signaling cascades that occur as cells sense confining forces and how that translates to behavioral changes, including elongated and multinucleated cell morphologies, novel migrational mechanisms, and altered gene expression, leading to a unique MSC secretome that could hold great promise for anti-inflammatory treatments. Through comparison of these altered behaviors, we aim to discern how MSCs alter their lineage selection, while tumor cells may become more aggressive and invasive. Synthesizing this information can be useful for employing MSCs for therapeutic approaches through systemic injections or tissue engineered grafts, and developing improved strategies for metastatic cancer therapies.
Collapse
Affiliation(s)
- Mary T. Doolin
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
| | - Rebecca A. Moriarty
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
- Maryland Biophysics Program, University of Maryland, College Park, College Park, MD, United States
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|
48
|
Bahcecioglu G, Basara G, Ellis BW, Ren X, Zorlutuna P. Breast cancer models: Engineering the tumor microenvironment. Acta Biomater 2020; 106:1-21. [PMID: 32045679 PMCID: PMC7185577 DOI: 10.1016/j.actbio.2020.02.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/14/2020] [Accepted: 02/05/2020] [Indexed: 12/24/2022]
Abstract
The mechanisms behind cancer initiation and progression are not clear. Therefore, development of clinically relevant models to study cancer biology and drug response in tumors is essential. In vivo models are very valuable tools for studying cancer biology and for testing drugs; however, they often suffer from not accurately representing the clinical scenario because they lack either human cells or a functional immune system. On the other hand, two-dimensional (2D) in vitro models lack the three-dimensional (3D) network of cells and extracellular matrix (ECM) and thus do not represent the tumor microenvironment (TME). As an alternative approach, 3D models have started to gain more attention, as such models offer a platform with the ability to study cell-cell and cell-material interactions parametrically, and possibly include all the components present in the TME. Here, we first give an overview of the breast cancer TME, and then discuss the current state of the pre-clinical breast cancer models, with a focus on the engineered 3D tissue models. We also highlight two engineering approaches that we think are promising in constructing models representative of human tumors: 3D printing and microfluidics. In addition to giving basic information about the TME in the breast tissue, this review article presents the state-of-the-art tissue engineered breast cancer models. STATEMENT OF SIGNIFICANCE: Involvement of biomaterials and tissue engineering fields in cancer research enables realistic mimicry of the cell-cell and cell-extracellular matrix (ECM) interactions in the tumor microenvironment (TME), and thus creation of better models that reflect the tumor response against drugs. Engineering the 3D in vitro models also requires a good understanding of the TME. Here, an overview of the breast cancer TME is given, and the current state of the pre-clinical breast cancer models, with a focus on the engineered 3D tissue models is discussed. This review article is useful not only for biomaterials scientists aiming to engineer 3D in vitro TME models, but also for cancer researchers willing to use these models for studying cancer biology and drug testing.
Collapse
Affiliation(s)
- Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Xiang Ren
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Pinar Zorlutuna
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, IN 46556, United States; Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, United States; Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, United States; Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46556, United States.
| |
Collapse
|
49
|
Pham QL, Tong A, Rodrigues LN, Zhao Y, Surblyte M, Ramos D, Brito J, Rahematpura A, Voronov RS. Ranking migration cue contributions to guiding individual fibroblasts faced with a directional decision in simple microfluidic bifurcations. Integr Biol (Camb) 2020; 11:208-220. [PMID: 31251334 DOI: 10.1093/intbio/zyz018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 04/04/2019] [Accepted: 05/21/2019] [Indexed: 01/02/2023]
Abstract
Directed cell migration in complex micro-environments, such as in vivo pores, is important for predicting locations of artificial tissue growth and optimizing scaffold architectures. Yet, the directional decisions of cells facing multiple physiochemical cues have not been characterized. Hence, we aim to provide a ranking of the relative importance of the following cues to the decision-making of individual fibroblast cells: chemoattractant concentration gradient, channel width, mitosis, and contact-guidance. In this study, bifurcated micro-channels with branches of different widths were created. Fibroblasts were then allowed to travel across these geometries by following a gradient of platelet-derived growth factor-BB (PDGF-BB) established inside the channels. Subsequently, a combination of statistical analysis and image-based diffusion modeling was used to report how the presence of multiple complex migration cues, including cell-cell influences, affect the fibroblast decision-making. It was found that the cells prefer wider channels over a higher chemoattractant gradient when choosing between asymmetric bifurcated branches. Only when the branches were symmetric in width did the gradient become predominant in directing which path the cell will take. Furthermore, when both the gradient and the channels were symmetric, contact guidance became important for guiding the cells in making directional choices. Based on these results we were able to rank these directional cues from most influential to the least as follows: mitosis > channel width asymmetry > chemoattractant gradient difference > and contact-guidance. It is expected that these results will benefit the fields of regenerative medicine, wound healing and developmental biology.
Collapse
Affiliation(s)
- Quang Long Pham
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Anh Tong
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Lydia N Rodrigues
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Yang Zhao
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Migle Surblyte
- Ying Wu College of Computing Sciences, Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, USA
| | - Diomar Ramos
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - John Brito
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Adwik Rahematpura
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Roman S Voronov
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| |
Collapse
|
50
|
Aggregation-induced emission luminogen for specific identification of malignant tumour in vivo. Sci China Chem 2020. [DOI: 10.1007/s11426-019-9677-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|