51
|
Hohmann U, Ghadban C, Prell J, Strauss C, Dehghani F, Hohmann T. A toolbox to analyze collective cell migration, proliferation and cellular organization simultaneously. Cell Adh Migr 2023; 17:1-11. [PMID: 37938930 PMCID: PMC10773533 DOI: 10.1080/19336918.2023.2276615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/19/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Analyses of collective cell migration and orientation phenomena are needed to assess the behavior of multicellular clusters. While some tools to the authors' knowledge none is capable to analyze collective migration, cellular orientation and proliferation in phase contrast images simultaneously. METHODS We provide a tool based to analyze phase contrast images of dense cell layers. PIV is used to calculatevelocity fields, while the structure tensor provides cellular orientation. An artificial neural network is used to identify cell division events, allowing to correlate migratory and organizational phenomena with cell density. CONCLUSION The presented tool allows the simultaneous analysis of collective cell behavior from phase contrast images in terms of migration, (self-)organization and proliferation.
Collapse
Affiliation(s)
- Urszula Hohmann
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chalid Ghadban
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Julian Prell
- Department of Neurosurgery, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christian Strauss
- Department of Neurosurgery, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Tim Hohmann
- Department of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
52
|
Melo S, Guerrero P, Moreira Soares M, Bordin JR, Carneiro F, Carneiro P, Dias MB, Carvalho J, Figueiredo J, Seruca R, Travasso RDM. The ECM and tissue architecture are major determinants of early invasion mediated by E-cadherin dysfunction. Commun Biol 2023; 6:1132. [PMID: 37938268 PMCID: PMC10632478 DOI: 10.1038/s42003-023-05482-x] [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: 01/20/2023] [Accepted: 10/18/2023] [Indexed: 11/09/2023] Open
Abstract
Germline mutations of E-cadherin cause Hereditary Diffuse Gastric Cancer (HDGC), a highly invasive cancer syndrome characterised by the occurrence of diffuse-type gastric carcinoma and lobular breast cancer. In this disease, E-cadherin-defective cells are detected invading the adjacent stroma since very early stages. Although E-cadherin loss is well established as a triggering event, other determinants of the invasive process persist largely unknown. Herein, we develop an experimental strategy that comprises in vitro extrusion assays using E-cadherin mutants associated to HDGC, as well as mathematical models epitomising epithelial dynamics and its interaction with the extracellular matrix (ECM). In vitro, we verify that E-cadherin dysfunctional cells detach from the epithelial monolayer and extrude basally into the ECM. Through phase-field modelling we demonstrate that, aside from loss of cell-cell adhesion, increased ECM attachment further raises basal extrusion efficiency. Importantly, by combining phase-field and vertex model simulations, we show that the cylindrical structure of gastric glands strongly promotes the cell's invasive ability. Moreover, we validate our findings using a dissipative particle dynamics simulation of epithelial extrusion. Overall, we provide the first evidence that cancer cell invasion is the outcome of defective cell-cell linkages, abnormal interplay with the ECM, and a favourable 3D tissue structure.
Collapse
Affiliation(s)
- Soraia Melo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, Porto, Portugal
| | - Pilar Guerrero
- Departamento de Matemáticas and Grupo Interdisciplinar de Sistemas Complejos (GISC), Universidad Carlos III de Madrid, Leganés, Spain
| | - Maurício Moreira Soares
- Oslo Center for Biostatistics and Epidemiology, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - José Rafael Bordin
- Department of Physics, Institute of Physics and Mathematics, Federal University of Pelotas, Capão do Leão, Rio Grande do Sul, Brazil
| | - Fátima Carneiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Patrícia Carneiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, Porto, Portugal
| | - Maria Beatriz Dias
- CISUC, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
| | - João Carvalho
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Joana Figueiredo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, Porto, Portugal.
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Raquel Seruca
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Ipatimup - Institute of Molecular Pathology and Immunology of the University of Porto, University of Porto, Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rui D M Travasso
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
53
|
Tzeng YDT, Hsiao JH, Tseng LM, Hou MF, Li CJ. Breast cancer organoids derived from patients: A platform for tailored drug screening. Biochem Pharmacol 2023; 217:115803. [PMID: 37709150 DOI: 10.1016/j.bcp.2023.115803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
Breast cancer stands as the most prevalent and heterogeneous malignancy affecting women globally, posing a substantial health concern. Enhanced comprehension of tumor pathology and the development of novel therapeutics are pivotal for advancing breast cancer treatment. Contemporary breast cancer investigation heavily leans on in vivo models and conventional cell culture techniques. Nonetheless, these approaches often encounter high failure rates in clinical trials due to species disparities and tissue structure variations. To address this, three-dimensional cultivation of organoids, resembling organ-like structures, has emerged as a promising alternative. Organoids represent innovative in vitro models that mirror in vivo tissue microenvironments. They retain the original tumor's diversity and facilitate the expansion of tumor samples from diverse origins, facilitating the representation of varying tumor stages. Optimized breast cancer organoid models, under precise culture conditions, offer benefits including convenient sample acquisition, abbreviated cultivation durations, and genetic stability. These attributes ensure a faithful replication of in vivo traits of breast cancer cells. As intricate cellular entities boasting spatial arrangements, breast cancer organoid models harbor substantial potential in precision medicine, organ transplantation, modeling intricate diseases, gene therapy, and drug innovation. This review delivers an overview of organoid culture techniques and outlines future prospects for organoid modeling.
Collapse
Affiliation(s)
- Yen-Dun Tony Tzeng
- Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Jui-Hu Hsiao
- Department of Surgery, Kaohsiung Municipal Minsheng Hospital, Kaohsiung, Taiwan
| | - Ling-Ming Tseng
- School of Medicine, National Yang-Ming University, Taipei 112, Taiwan; Comprehensive Breast Health Center, Taipei Veterans General Hospital, Taipei 112, Taiwan.
| | - Ming-Feng Hou
- Division of Breast Surgery, Department of Surgery, Center for Cancer Research, Kaohsiung Medical University Chung-Ho Memorial Hospital, Kaohsiung 807, Taiwan.
| | - Chia-Jung Li
- Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan; Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
54
|
Schab AM, Greenwade MM, Stock E, Lomonosova E, Cho K, Grither WR, Noia H, Wilke D, Mullen MM, Hagemann AR, Hagemann IS, Thaker PH, Kuroki LM, McCourt CK, Khabele D, Powell MA, Mutch DG, Zhao P, Shriver LP, Patti GJ, Longmore GD, Fuh KC. Stromal DDR2 Promotes Ovarian Cancer Metastasis through Regulation of Metabolism and Secretion of Extracellular Matrix Proteins. Mol Cancer Res 2023; 21:1234-1248. [PMID: 37527178 PMCID: PMC10832402 DOI: 10.1158/1541-7786.mcr-23-0347] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/03/2023] [Accepted: 07/28/2023] [Indexed: 08/03/2023]
Abstract
Ovarian cancer is the leading cause of gynecologic cancer-related deaths. The propensity for metastasis within the peritoneal cavity is a driving factor for the poor outcomes associated with this disease, but there is currently no effective therapy targeting metastasis. In this study, we investigate the contribution of stromal cells to ovarian cancer metastasis and identify normal stromal cell expression of the collagen receptor, discoidin domain receptor 2 (DDR2), that acts to facilitate ovarian cancer metastasis. In vivo, global genetic inactivation of Ddr2 impairs the ability of Ddr2-expressing syngeneic ovarian cancer cells to spread throughout the peritoneal cavity. Specifically, DDR2 expression in mesothelial cells lining the peritoneal cavity facilitates tumor cell attachment and clearance. Subsequently, omentum fibroblast expression of DDR2 promotes tumor cell invasion. Mechanistically, we find DDR2-expressing fibroblasts are more energetically active, such that DDR2 regulates glycolysis through AKT/SNAI1 leading to suppressed fructose-1,6-bisphosphatase and increased hexokinase activity, a key glycolytic enzyme. Upon inhibition of DDR2, we find decreased protein synthesis and secretion. Consequently, when DDR2 is inhibited, there is reduction in secreted extracellular matrix proteins important for metastasis. Specifically, we find that fibroblast DDR2 inhibition leads to decreased secretion of the collagen crosslinker, LOXL2. Adding back LOXL2 to DDR2 deficient fibroblasts rescues the ability of tumor cells to invade. Overall, our results suggest that stromal cell expression of DDR2 is an important mediator of ovarian cancer metastasis. IMPLICATIONS DDR2 is highly expressed by stromal cells in ovarian cancer that can mediate metastasis and is a potential therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Angela M. Schab
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Molly M. Greenwade
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Elizabeth Stock
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Elena Lomonosova
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Kevin Cho
- Center for Metabolomics and Isotope Tracing, Department of Chemistry, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Whitney R. Grither
- Department of Obstetrics and Gynecology, Barnes Jewish Hospital, Washington University, St. Louis, MO 63110, USA
| | - Hollie Noia
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Daniel Wilke
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Mary M. Mullen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Andrea R. Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Ian S. Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Premal H. Thaker
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Lindsay M. Kuroki
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Carolyn K. McCourt
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Dineo Khabele
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
| | - Matthew A. Powell
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - David G. Mutch
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
| | - Peinan Zhao
- Department of Obstetrics and Gynecology, Barnes Jewish Hospital, Washington University, St. Louis, MO 63110, USA
| | - Leah P. Shriver
- Center for Metabolomics and Isotope Tracing, Department of Chemistry, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Gary J. Patti
- Center for Metabolomics and Isotope Tracing, Department of Chemistry, Department of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Gregory D. Longmore
- Division of Oncology, Department of Medicine Washington University, St. Louis. MO 63110, USA
- ICCE Institute, Washington University, St. Louis MO 63110, USA
| | - Katherine C. Fuh
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, St. Louis, MO 63110, USA
- Center for Reproductive Health Sciences, Division of Biology and Biomedical Sciences, Washington University, St. Louis, MO 63110, USA
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology University of California, San Francisco, San Francisco, CA 94143 USA
| |
Collapse
|
55
|
Xin Y, Li K, Huang M, Liang C, Siemann D, Wu L, Tan Y, Tang X. Biophysics in tumor growth and progression: from single mechano-sensitive molecules to mechanomedicine. Oncogene 2023; 42:3457-3490. [PMID: 37864030 PMCID: PMC10656290 DOI: 10.1038/s41388-023-02844-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 09/08/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Evidence from physical sciences in oncology increasingly suggests that the interplay between the biophysical tumor microenvironment and genetic regulation has significant impact on tumor progression. Especially, tumor cells and the associated stromal cells not only alter their own cytoskeleton and physical properties but also remodel the microenvironment with anomalous physical properties. Together, these altered mechano-omics of tumor tissues and their constituents fundamentally shift the mechanotransduction paradigms in tumorous and stromal cells and activate oncogenic signaling within the neoplastic niche to facilitate tumor progression. However, current findings on tumor biophysics are limited, scattered, and often contradictory in multiple contexts. Systematic understanding of how biophysical cues influence tumor pathophysiology is still lacking. This review discusses recent different schools of findings in tumor biophysics that have arisen from multi-scale mechanobiology and the cutting-edge technologies. These findings range from the molecular and cellular to the whole tissue level and feature functional crosstalk between mechanotransduction and oncogenic signaling. We highlight the potential of these anomalous physical alterations as new therapeutic targets for cancer mechanomedicine. This framework reconciles opposing opinions in the field, proposes new directions for future cancer research, and conceptualizes novel mechanomedicine landscape to overcome the inherent shortcomings of conventional cancer diagnosis and therapies.
Collapse
Grants
- R35 GM150812 NIGMS NIH HHS
- This work was financially supported by National Natural Science Foundation of China (Project no. 11972316, Y.T.), Shenzhen Science and Technology Innovation Commission (Project no. JCYJ20200109142001798, SGDX2020110309520303, and JCYJ20220531091002006, Y.T.), General Research Fund of Hong Kong Research Grant Council (PolyU 15214320, Y. T.), Health and Medical Research Fund (HMRF18191421, Y.T.), Hong Kong Polytechnic University (1-CD75, 1-ZE2M, and 1-ZVY1, Y.T.), the Cancer Pilot Research Award from UF Health Cancer Center (X. T.), the National Institute of General Medical Sciences of the National Institutes of Health under award number R35GM150812 (X. T.), the National Science Foundation under grant number 2308574 (X. T.), the Air Force Office of Scientific Research under award number FA9550-23-1-0393 (X. T.), the University Scholar Program (X. T.), UF Research Opportunity Seed Fund (X. T.), the Gatorade Award (X. T.), and the National Science Foundation REU Site at UF: Engineering for Healthcare (Douglas Spearot and Malisa Sarntinoranont). We are deeply grateful for the insightful discussions with and generous support from all members of Tang (UF)’s and Tan (PolyU)’s laboratories and all staff members of the MAE/BME/ECE/Health Cancer Center at UF and BME at PolyU.
- National Natural Science Foundation of China (National Science Foundation of China)
- Shenzhen Science and Technology Innovation Commission
Collapse
Affiliation(s)
- Ying Xin
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Keming Li
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China
| | - Miao Huang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Chenyu Liang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
| | - Dietmar Siemann
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Lizi Wu
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Youhua Tan
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China.
- Research Institute of Smart Ageing, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Xin Tang
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA.
- UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
56
|
Abstract
Multicellular organisms generate tissues of diverse shapes and functions from cells and extracellular matrices. Their adhesion molecules mediate cell-cell and cell-matrix interactions, which not only play crucial roles in maintaining tissue integrity but also serve as key regulators of tissue morphogenesis. Cells constantly probe their environment to make decisions: They integrate chemical and mechanical information from the environment via diffusible ligand- or adhesion-based signaling to decide whether to release specific signaling molecules or enzymes, to divide or differentiate, to move away or stay, or even whether to live or die. These decisions in turn modify their environment, including the chemical nature and mechanical properties of the extracellular matrix. Tissue morphology is the physical manifestation of the remodeling of cells and matrices by their historical biochemical and biophysical landscapes. We review our understanding of matrix and adhesion molecules in tissue morphogenesis, with an emphasis on key physical interactions that drive morphogenesis.
Collapse
Affiliation(s)
- Di Wu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA;
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA;
| | - Shaohe Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA;
| |
Collapse
|
57
|
Gong X, Wen Z, Liang Z, Xiao H, Lee S, Wright T, Nguyen RY, Rossello A, Mak M. Instant Assembly of Collagen for Scaffolding, Tissue Engineering, and Bioprinting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.08.561456. [PMID: 37873099 PMCID: PMC10592672 DOI: 10.1101/2023.10.08.561456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Controllable assembly of cells and tissues offers potential for advancing disease and development modeling and regenerative medicine. The body's natural scaffolding material is the extracellular matrix, composed largely of collagen I. However, challenges in precisely controlling collagen assembly limit collagen's applicability as a primary bioink or glue for biofabrication. Here, we introduce a set of biopatterning methods, termed Tunable Rapid Assembly of Collagenous Elements (TRACE), that enables instant gelation and rapid patterning of collagen I solutions with wide range of concentrations. Our methods are based on accelerating the gelation of collagen solutions to instantaneous speeds via macromolecular crowding, allowing versatile patterning of both cell-free and cell-laden collagen-based bioinks. We demonstrate notable applications, including macroscopic organoid engineering, rapid free-form 3D bioprinting, contractile cardiac ventricle model, and patterning of high-resolution (below 5 (m) collagen filament. Our findings enable more controllable and versatile applications for multi-scale collagen-based biofabrication.
Collapse
|
58
|
Xu KL, Caprio ND, Fallahi H, Dehgany M, Davidson MD, Cheung BC, Laforest L, Wu M, Shenoy V, Han L, Mauck RL, Burdick JA. Microinterfaces in bicontinuous hydrogels guide rapid 3D cell migration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559609. [PMID: 37808836 PMCID: PMC10557715 DOI: 10.1101/2023.09.28.559609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Cell migration is critical for tissue development and regeneration but requires extracellular environments that are conducive to motion. Cells may actively generate migratory routes in vivo by degrading or remodeling their environments or may instead utilize existing ECM microstructures or microtracks as innate pathways for migration. While hydrogels in general are valuable tools for probing the extracellular regulators of 3D migration, few have recapitulated these natural migration paths. Here, we developed a biopolymer-based (i.e., gelatin and hyaluronic acid) bicontinuous hydrogel system formed through controlled solution immiscibility whose continuous subdomains and high micro-interfacial surface area enabled rapid 3D migration, particularly when compared to homogeneous hydrogels. Migratory behavior was mesenchymal in nature and regulated by biochemical and biophysical signals from the hydrogel, which was shown across various cell types and physiologically relevant contexts (e.g., cell spheroids, ex vivo tissues, in vivo tissues). Our findings introduce a new design that leverages important local interfaces to guide rapid cell migration.
Collapse
|
59
|
Castro-Abril H, Heras J, Del Barrio J, Paz L, Alcaine C, Aliácar MP, Garzón-Alvarado D, Doblaré M, Ochoa I. The Role of Mechanical Properties and Structure of Type I Collagen Hydrogels on Colorectal Cancer Cell Migration. Macromol Biosci 2023; 23:e2300108. [PMID: 37269065 DOI: 10.1002/mabi.202300108] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/05/2023] [Indexed: 06/04/2023]
Abstract
Mechanical interactions between cells and their microenvironment play an important role in determining cell fate, which is particularly relevant in metastasis, a process where cells invade tissue matrices with different mechanical properties. In vitro, type I collagen hydrogels have been commonly used for modeling the microenvironment due to its ubiquity in the human body. In this work, the combined influence of the stiffness of these hydrogels and their ultrastructure on the migration patterns of HCT-116 and HT-29 spheroids are analyzed. For this, six different types of pure type I collagen hydrogels by changing the collagen concentration and the gelation temperature are prepared. The stiffness of each sample is measured and its ultrastructure is characterized. Cell migration studies are then performed by seeding the spheroids in three different spatial conditions. It is shown that changes in the aforementioned parameters lead to differences in the mechanical stiffness of the matrices as well as the ultrastructure. These differences, in turn, lead to distinct cell migration patterns of HCT-116 and HT-29 spheroids in either of the spatial conditions tested. Based on these results, it is concluded that the stiffness and the ultrastructural organization of the matrix can actively modulate cell migration behavior in colorectal cancer spheroids.
Collapse
Affiliation(s)
- Hector Castro-Abril
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Biomimetics Lab, National University of Colombia, Bogotá, 111321, Colombia
| | - Jónathan Heras
- Grupo de Informática, University of La Rioja, La Rioja, 26006, Spain
| | - Jesús Del Barrio
- Instituto de Nanociencia y Materiales de Aragón (INMA), Department of Organic Chemistry, CSIC-University of Zaragoza, Zaragoza, 50018, Spain
| | - Laura Paz
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, 50018, Spain
| | - Clara Alcaine
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
| | - Marina Pérez Aliácar
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, 50018, Spain
| | | | - Manuel Doblaré
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, 50018, Spain
- Nanjing Tech University, Nanjing, 50018, China
| | - Ignacio Ochoa
- Tissue Microenvironment lab (TME lab), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain
- Instituto de Investigación Sanitaria Aragón (IISA), Zaragoza, 50018, Spain
- Centro Investigación Biomédica en Red. Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, 50018, Spain
- Nanjing Tech University, Nanjing, 50018, China
| |
Collapse
|
60
|
Su CY, Matsubara T, Wu A, Ahn EH, Kim DH. Matrix Anisotropy Promotes a Transition of Collective to Disseminated Cell Migration via a Collective Vortex Motion. Adv Biol (Weinh) 2023; 7:e2300026. [PMID: 36932886 DOI: 10.1002/adbi.202300026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Indexed: 03/19/2023]
Abstract
Cells detached and disseminated away from collectively migrating cells are frequently found during tumor invasion at the invasion front, where extracellular matrix (ECM) fibers are parallel to the cell migration direction. However, it remains unclear how anisotropic topography promotes the transition of collective to disseminated cell migration. This study applies a collective cell migration model with and without 800 nm wide aligned nanogrooves parallel, perpendicular, or diagonal to the cell migration direction. After 120 hour migration, MCF7-GFP-H2B-mCherry breast cancer cells display more disseminated cells at the migration front on parallel topography than on other topographies. Notably, a fluid-like collective motion with high vorticity is enhanced at the migration front on parallel topography. Furthermore, high vorticity but not velocity is correlated with disseminated cell numbers on parallel topography. Enhanced collective vortex motion colocalizes with cell monolayer defects where cells extend protrusions into the free space, suggesting that topography-driven cell crawling for defect closure promotes the collective vortex motion. In addition, elongated cell morphology and frequent protrusions induced by topography may further contribute to the collective vortex motion. Overall, a high-vorticity collective motion at the migration front promoted by parallel topography suggests a cause of the transition of collective to disseminated cell migration.
Collapse
Affiliation(s)
- Chia-Yi Su
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Tatsuya Matsubara
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Alex Wu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Eun Hyun Ahn
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
61
|
Joshi A, Singh N. Generation of Patterned Cocultures in 2D and 3D: State of the Art. ACS OMEGA 2023; 8:34249-34261. [PMID: 37780002 PMCID: PMC10536108 DOI: 10.1021/acsomega.3c02713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/30/2023] [Indexed: 10/03/2023]
Abstract
Cells inside the body are embedded into a highly structured microenvironment that consists of cells that lie in direct or close contact with other cell types that regulate the overall tissue function. Therefore, coculture models are versatile tools that can generate tissue engineering constructs with improved mimicking of in vivo conditions. While there are many reviews that have focused on pattering a single cell type, very few reviews have been focused on techniques for coculturing multiple cell types on a single substrate with precise control. In this regard, this Review covers various technologies that have been utilized for the development of these patterned coculture models while mentioning the limitations associated with each of them. Further, the application of these models to various tissue engineering applications has been discussed.
Collapse
Affiliation(s)
- Akshay Joshi
- Centre
for Biomedical Engineering, Indian Institute
of Technology Delhi, Hauz Khas, New Delhi, Delhi 110016, India
| | - Neetu Singh
- Centre
for Biomedical Engineering, Indian Institute
of Technology Delhi, Hauz Khas, New Delhi, Delhi 110016, India
- Biomedical
Engineering Unit, All India Institute of
Medical Sciences, Ansari
Nagar, New Delhi, Delhi 110029, India
| |
Collapse
|
62
|
Séraudie I, Pillet C, Cesana B, Bazelle P, Jeanneret F, Evrard B, Chalmel F, Bouzit A, Battail C, Long JA, Descotes JL, Cochet C, Filhol O. A new scaffold-free tumoroid model provides a robust preclinical tool to investigate invasion and drug response in Renal Cell Carcinoma. Cell Death Dis 2023; 14:622. [PMID: 37736770 PMCID: PMC10517165 DOI: 10.1038/s41419-023-06133-z] [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: 04/12/2023] [Revised: 08/24/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023]
Abstract
Clear cell Renal Cell Carcinoma (ccRCC) is one of the most prevalent kidney cancers, which is often asymptomatic and thus discovered at a metastatic state (mRCC). mRCC are highly heterogeneous tumors composed of subclonal populations that lead to poor treatment response rate. Several recent works explored the potential of ccRCC tumoroids culture derived from patients. However, these models were produced following a scaffold-based method using collagen I or Matrigel that exhibit lot variability and whose complexity could induce treatment response modifications and phenotypic alterations. Following the observation that ccRCC tumoroids can create their own niche by secreting extracellular matrix components, we developed the first scaffold-free tumoroid model of ccRCC tumors. Tumoroids from mice as well as from human tumors were generated with high success rate (≥90%) using a magnetic suspension method and standard culture media. Immunofluorescence analysis revealed their self-organization capacities to maintain multiple tumor-resident cell types, including endothelial progenitor cells. Transcriptomic analysis showed the reproducibility of the method highlighting that the majority of gene expression patterns was conserved in tumoroids compared to their matching tumor tissue. Moreover, this model enables to evaluate drug effects and invasiveness of renal cancer cells in a 3D context, providing a robust preclinical tool for drug screening and biomarker assessment in line with alternative ex vivo methods like tumor tissue slice culture or in vivo xenograft models.
Collapse
Grants
- EC | Horizon 2020 Framework Programme (EU Framework Programme for Research and Innovation H2020)
- INSERM, CEA, Ligue Comité de l’Isère, University Grenoble Alpes, Centre Hospitalier Universitaire de Grenoble-Alpes (CHUGA), Groupement des Entreprises Françaises dans la Lutte contre le Cancer (GEFLUC)
- CEA, UGA
- CEA, Inserm
- UGA, Inserm, CEA
- CHU, Ligue Comité de l’Isère
- Inserm, Ligue Comité de l’Isère
Collapse
Affiliation(s)
- Irinka Séraudie
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Catherine Pillet
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Beatrice Cesana
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Pauline Bazelle
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Florian Jeanneret
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UA 13, F-38000, Grenoble, France
| | - Bertrand Evrard
- University Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Frédéric Chalmel
- University Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, F-35000, Rennes, France
| | - Assilah Bouzit
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043, Grenoble, cedex 9, France
| | - Christophe Battail
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UA 13, F-38000, Grenoble, France
| | - Jean-Alexandre Long
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043, Grenoble, cedex 9, France
| | - Jean Luc Descotes
- Centre hospitalier universitaire Grenoble Alpes, CS 10217, 38043, Grenoble, cedex 9, France
| | - Claude Cochet
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France
| | - Odile Filhol
- University Grenoble Alpes, Inserm, CEA, IRIG-Biosanté, UMR 1292, F-38000, Grenoble, France.
| |
Collapse
|
63
|
Lin W, Fang J, Wei S, He G, Liu J, Li X, Peng X, Li D, Yang S, Li X, Yang L, Li H. Extracellular vesicle-cell adhesion molecules in tumours: biofunctions and clinical applications. Cell Commun Signal 2023; 21:246. [PMID: 37735659 PMCID: PMC10512615 DOI: 10.1186/s12964-023-01236-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/18/2023] [Indexed: 09/23/2023] Open
Abstract
Cell adhesion molecule (CAM) is an umbrella term for several families of molecules, including the cadherin family, integrin family, selectin family, immunoglobulin superfamily, and some currently unclassified adhesion molecules. Extracellular vesicles (EVs) are important information mediators in cell-to-cell communication. Recent evidence has confirmed that CAMs transported by EVs interact with recipient cells to influence EV distribution in vivo and regulate multiple cellular processes. This review focuses on the loading of CAMs onto EVs, the roles of CAMs in regulating EV distribution, and the known and possible mechanisms of these actions. Moreover, herein, we summarize the impacts of CAMs transported by EVs to the tumour microenvironment (TME) on the malignant behaviour of tumour cells (proliferation, metastasis, immune escape, and so on). In addition, from the standpoint of clinical applications, the significance and challenges of using of EV-CAMs in the diagnosis and therapy of tumours are discussed. Finally, considering recent advances in the understanding of EV-CAMs, we outline significant challenges in this field that require urgent attention to advance research and promote the clinical applications of EV-CAMs. Video Abstract.
Collapse
Affiliation(s)
- Weikai Lin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Jianjun Fang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xian Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Dai Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Shenyang Clinical Medical Research Center for Diagnosis, Treatment and Health Management of Early Digestive Cancer, Shenyang, 110032, China.
| |
Collapse
|
64
|
Sauer F, Grosser S, Shahryari M, Hayn A, Guo J, Braun J, Briest S, Wolf B, Aktas B, Horn L, Sack I, Käs JA. Changes in Tissue Fluidity Predict Tumor Aggressiveness In Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303523. [PMID: 37553780 PMCID: PMC10502644 DOI: 10.1002/advs.202303523] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Indexed: 08/10/2023]
Abstract
Cancer progression is caused by genetic changes and associated with various alterations in cell properties, which also affect a tumor's mechanical state. While an increased stiffness has been well known for long for solid tumors, it has limited prognostic power. It is hypothesized that cancer progression is accompanied by tissue fluidization, where portions of the tissue can change position across different length scales. Supported by tabletop magnetic resonance elastography (MRE) on stroma mimicking collagen gels and microscopic analysis of live cells inside patient derived tumor explants, an overview is provided of how cancer associated mechanisms, including cellular unjamming, proliferation, microenvironment composition, and remodeling can alter a tissue's fluidity and stiffness. In vivo, state-of-the-art multifrequency MRE can distinguish tumors from their surrounding host tissue by their rheological fingerprints. Most importantly, a meta-analysis on the currently available clinical studies is conducted and universal trends are identified. The results and conclusions are condensed into a gedankenexperiment about how a tumor can grow and eventually metastasize into its environment from a physics perspective to deduce corresponding mechanical properties. Based on stiffness, fluidity, spatial heterogeneity, and texture of the tumor front a roadmap for a prognosis of a tumor's aggressiveness and metastatic potential is presented.
Collapse
Affiliation(s)
- Frank Sauer
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
| | - Steffen Grosser
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
- Institute for Bioengineering of CataloniaThe Barcelona Institute for Science and Technology (BIST)Barcelona08028Spain
| | - Mehrgan Shahryari
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Alexander Hayn
- Department of HepatologyLeipzig University Hospital04103LeipzigGermany
| | - Jing Guo
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Jürgen Braun
- Institute of Medical InformaticsCharité‐Universitätsmedizin10117BerlinGermany
| | - Susanne Briest
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Benjamin Wolf
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Bahriye Aktas
- Department of GynecologyLeipzig University Hospital04103LeipzigGermany
| | - Lars‐Christian Horn
- Division of Breast, Urogenital and Perinatal PathologyLeipzig University Hospital04103LeipzigGermany
| | - Ingolf Sack
- Department of RadiologyCharité‐Universitätsmedizin10117BerlinGermany
| | - Josef A. Käs
- Soft Matter Physics DivisionPeter‐Debye‐Institute for Soft Matter Physics04103LeipzigGermany
| |
Collapse
|
65
|
Palmiero M, Cantarosso I, di Blasio L, Monica V, Peracino B, Primo L, Puliafito A. Collective directional migration drives the formation of heteroclonal cancer cell clusters. Mol Oncol 2023; 17:1699-1725. [PMID: 36587372 PMCID: PMC10483614 DOI: 10.1002/1878-0261.13369] [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: 08/09/2022] [Revised: 11/11/2022] [Accepted: 12/30/2022] [Indexed: 01/02/2023] Open
Abstract
Metastasisation occurs through the acquisition of invasive and survival capabilities that allow tumour cells to colonise distant sites. While the role of multicellular aggregates in cancer dissemination is acknowledged, the mechanisms that drive the formation of multiclonal cell aggregates are not fully elucidated. Here, we show that cancer cells of different tissue of origins can perform collective directional migration and can actively form heteroclonal aggregates in 3D, through a proliferation-independent mechanism. Coalescence of distant cell clusters is mediated by subcellular actin-rich protrusions and multicellular outgrowths that extend towards neighbouring aggregates. Coherently, perturbation of cytoskeletal dynamics impairs collective migration while myosin II activation is necessary for multicellular movements. We put forward the hypothesis that cluster attraction is mediated by secreted soluble factors. Such a hypothesis is consistent with the abrogation of aggregation by inhibition of PI3K/AKT/mTOR and MEK/ERK, the chemoattracting activity of conditioned culture media and with a wide screening of secreted proteins. Our results present a novel collective migration model and shed light on the mechanisms of formation of heteroclonal aggregates in cancer.
Collapse
Affiliation(s)
- Miriam Palmiero
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Isabel Cantarosso
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Laura di Blasio
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Valentina Monica
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Barbara Peracino
- Department of Clinical and Biological SciencesSan Luigi Hospital, University of TurinOrbassanoItaly
| | - Luca Primo
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| | - Alberto Puliafito
- Candiolo Cancer Institute, FPO – IRCCSCandioloItaly
- Department of OncologyUniversity of TurinCandioloItaly
| |
Collapse
|
66
|
Le HA, Mayor R. Cell-matrix and cell-cell interaction mechanics in guiding migration. Biochem Soc Trans 2023; 51:1733-1745. [PMID: 37610008 PMCID: PMC10586762 DOI: 10.1042/bst20230211] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/24/2023]
Abstract
Physical properties of tissue are increasingly recognised as major regulatory cues affecting cell behaviours, particularly cell migration. While these properties of the extracellular matrix have been extensively discussed, the contribution from the cellular components that make up the tissue are still poorly appreciated. In this mini-review, we will discuss two major physical components: stiffness and topology with a stronger focus on cell-cell interactions and how these can impact cell migration.
Collapse
Affiliation(s)
- Hoang Anh Le
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, U.K
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, U.K
| |
Collapse
|
67
|
Lee YL, Mathur J, Walter C, Zmuda H, Pathak A. Matrix obstructions cause multiscale disruption in collective epithelial migration by suppressing leader cell function. Mol Biol Cell 2023; 34:ar94. [PMID: 37379202 PMCID: PMC10398892 DOI: 10.1091/mbc.e22-06-0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 06/06/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023] Open
Abstract
During disease and development, physical changes in extracellular matrix cause jamming, unjamming, and scattering in epithelial migration. However, whether disruptions in matrix topology alter collective cell migration speed and cell-cell coordination remains unclear. We microfabricated substrates with stumps of defined geometry, density, and orientation, which create obstructions for migrating epithelial cells. Here, we show that cells lose their speed and directionality when moving through densely spaced obstructions. Although leader cells are stiffer than follower cells on flat substrates, dense obstructions cause overall cell softening. Through a lattice-based model, we identify cellular protrusions, cell-cell adhesions, and leader-follower communication as key mechanisms for obstruction-sensitive collective cell migration. Our modeling predictions and experimental validations show that cells' obstruction sensitivity requires an optimal balance of cell-cell adhesions and protrusions. Both MDCK (more cohesive) and α-catenin-depleted MCF10A cells were less obstruction sensitive than wild-type MCF10A cells. Together, microscale softening, mesoscale disorder, and macroscale multicellular communication enable epithelial cell populations to sense topological obstructions encountered in challenging environments. Thus, obstruction-sensitivity could define "mechanotype" of cells that collectively migrate yet maintain intercellular communication.
Collapse
Affiliation(s)
- Ye Lim Lee
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Christopher Walter
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| | - Hannah Zmuda
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130
- Department of Mechanical Engineering & Materials Science, Washington University in St. Louis, St. Louis, MO 63130
| |
Collapse
|
68
|
Saha S, Müller D, Clark AG. Mechanosensory feedback loops during chronic inflammation. Front Cell Dev Biol 2023; 11:1225677. [PMID: 37492225 PMCID: PMC10365287 DOI: 10.3389/fcell.2023.1225677] [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: 05/19/2023] [Accepted: 06/27/2023] [Indexed: 07/27/2023] Open
Abstract
Epithelial tissues are crucial to maintaining healthy organization and compartmentalization in various organs and act as a first line of defense against infection in barrier organs such as the skin, lungs and intestine. Disruption or injury to these barriers can lead to infiltration of resident or foreign microbes, initiating local inflammation. One often overlooked aspect of this response is local changes in tissue mechanics during inflammation. In this mini-review, we summarize known molecular mechanisms linking disruption of epithelial barrier function to mechanical changes in epithelial tissues. We consider direct mechanisms, such as changes in the secretion of extracellular matrix (ECM)-modulating enzymes by immune cells as well as indirect mechanisms including local activation of fibroblasts. We discuss how these mechanical changes can modulate local immune cell activity and inflammation and perturb epithelial homeostasis, further dysregulating epithelial barrier function. We propose that this two-way relationship between loss of barrier function and altered tissue mechanics can lead to a positive feedback loop that further perpetuates inflammation. We discuss this cycle in the context of several chronic inflammatory diseases, including inflammatory bowel disease (IBD), liver disease and cancer, and we present the modulation of tissue mechanics as a new framework for combating chronic inflammation.
Collapse
Affiliation(s)
- Sarbari Saha
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart, Germany
- University of Stuttgart, Stuttgart Research Center Systems Biology, Stuttgart, Germany
- University of Tübingen, Center for Personalized Medicine, Tübingen, Germany
| | - Dafne Müller
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart, Germany
| | - Andrew G. Clark
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart, Germany
- University of Stuttgart, Stuttgart Research Center Systems Biology, Stuttgart, Germany
- University of Tübingen, Center for Personalized Medicine, Tübingen, Germany
| |
Collapse
|
69
|
Srivastava V, Hu JL, Garbe JC, Veytsman B, Shalabi SF, Yllanes D, Thomson M, LaBarge MA, Huber G, Gartner ZJ. Configurational entropy is an intrinsic driver of tissue structural heterogeneity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.01.546933. [PMID: 37425903 PMCID: PMC10327153 DOI: 10.1101/2023.07.01.546933] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Tissues comprise ordered arrangements of cells that can be surprisingly disordered in their details. How the properties of single cells and their microenvironment contribute to the balance between order and disorder at the tissue-scale remains poorly understood. Here, we address this question using the self-organization of human mammary organoids as a model. We find that organoids behave like a dynamic structural ensemble at the steady state. We apply a maximum entropy formalism to derive the ensemble distribution from three measurable parameters - the degeneracy of structural states, interfacial energy, and tissue activity (the energy associated with positional fluctuations). We link these parameters with the molecular and microenvironmental factors that control them to precisely engineer the ensemble across multiple conditions. Our analysis reveals that the entropy associated with structural degeneracy sets a theoretical limit to tissue order and provides new insight for tissue engineering, development, and our understanding of disease progression.
Collapse
Affiliation(s)
- Vasudha Srivastava
- Dept. of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
| | - Jennifer L. Hu
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley, CA 94720, USA
| | - James C. Garbe
- Dept. of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Boris Veytsman
- Chan Zuckerberg Initiative, Redwood City, CA 94963, USA
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA
| | | | - David Yllanes
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Instituto de Biocomputaciòn y Fìsica de Sistemas Complejos (BIFI), 50018 Zaragoza, Spain
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mark A. LaBarge
- Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Greg Huber
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Zev J. Gartner
- Dept. of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA 94158, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Center for Cellular Construction, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
70
|
Parisi DR, Wiebke LE, Mandl JN, Textor J. Flow rate resonance of actively deforming particles. Sci Rep 2023; 13:9455. [PMID: 37301896 DOI: 10.1038/s41598-023-36182-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Lymphoid organs are unusual multicellular tissues: they are densely packed, but the lymphocytes trafficking through them are actively moving. We hypothesize that the intriguing ability of lymphocytes to avoid jamming and clogging is in part attributable to the dynamic shape changes that cells undergo when they move. In this work, we test this hypothesis by investigating an idealized system, namely, the flow of self-propelled, oscillating particles passing through a narrow constriction in two dimensions (2D), using numerical simulations. We found that deformation allows particles with these properties to flow through a narrow constriction in conditions when non-deformable particles would not be able to do so. Such a flowing state requires the amplitude and frequency of oscillations to exceed threshold values. Moreover, a resonance leading to the maximum flow rate was found when the oscillation frequency matched the natural frequency of the particle related to its elastic stiffness. To our knowledge, this phenomenon has not been described previously. Our findings could have important implications for understanding and controlling flow in a variety of systems in addition to lymphoid organs, such as granular flows subjected to vibration.
Collapse
Affiliation(s)
- Daniel R Parisi
- Instituto Tecnológico de Buenos Aires (ITBA), CONICET, C.A. de Buenos Aires, Argentina.
| | - Lucas E Wiebke
- Instituto Tecnológico de Buenos Aires (ITBA), C.A. de Buenos Aires, Argentina
| | - Judith N Mandl
- Department of Physiology and McGill Research Centre on Complex Traits, McGill University, Montreal, Canada
| | - Johannes Textor
- Data Science group, Institute for Computing and Information Sciences, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
71
|
Leartprapun N, Zeng Z, Hajjarian Z, Bossuyt V, Nadkarni SK. Speckle rheological spectroscopy reveals wideband viscoelastic spectra of biological tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544037. [PMID: 37333220 PMCID: PMC10274797 DOI: 10.1101/2023.06.08.544037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Mechanical transformation of tissue is not merely a symptom but a decisive driver in pathological processes. Comprising intricate network of cells, fibrillar proteins, and interstitial fluid, tissues exhibit distinct solid-(elastic) and liquid-like (viscous) behaviours that span a wide band of frequencies. Yet, characterization of wideband viscoelastic behaviour in whole tissue has not been investigated, leaving a vast knowledge gap in the higher frequency range that is linked to fundamental intracellular processes and microstructural dynamics. Here, we present wideband Speckle rHEologicAl spectRoScopy (SHEARS) to address this need. We demonstrate, for the first time, analysis of frequency-dependent elastic and viscous moduli up to the sub-MHz regime in biomimetic scaffolds and tissue specimens of blood clots, breast tumours, and bone. By capturing previously inaccessible viscoelastic behaviour across the wide frequency spectrum, our approach provides distinct and comprehensive mechanical signatures of tissues that may provide new mechanobiological insights and inform novel disease prognostication.
Collapse
Affiliation(s)
- Nichaluk Leartprapun
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Ziqian Zeng
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Zeinab Hajjarian
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| | - Veerle Bossuyt
- Department of Pathology, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Seemantini K. Nadkarni
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
72
|
Dow LP, Parmar T, Marchetti MC, Pruitt BL. Engineering tools for quantifying and manipulating forces in epithelia. BIOPHYSICS REVIEWS 2023; 4:021303. [PMID: 38510344 PMCID: PMC10903508 DOI: 10.1063/5.0142537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 04/20/2023] [Indexed: 03/22/2024]
Abstract
The integrity of epithelia is maintained within dynamic mechanical environments during tissue development and homeostasis. Understanding how epithelial cells mechanosignal and respond collectively or individually is critical to providing insight into developmental and (patho)physiological processes. Yet, inferring or mimicking mechanical forces and downstream mechanical signaling as they occur in epithelia presents unique challenges. A variety of in vitro approaches have been used to dissect the role of mechanics in regulating epithelia organization. Here, we review approaches and results from research into how epithelial cells communicate through mechanical cues to maintain tissue organization and integrity. We summarize the unique advantages and disadvantages of various reduced-order model systems to guide researchers in choosing appropriate experimental systems. These model systems include 3D, 2D, and 1D micromanipulation methods, single cell studies, and noninvasive force inference and measurement techniques. We also highlight a number of in silico biophysical models that are informed by in vitro and in vivo observations. Together, a combination of theoretical and experimental models will aid future experiment designs and provide predictive insight into mechanically driven behaviors of epithelial dynamics.
Collapse
Affiliation(s)
| | - Toshi Parmar
- Department of Physics, University of California Santa Barbara, Santa Barbara, California 93106, USA
| | | | | |
Collapse
|
73
|
Sievers J, Mahajan V, Welzel PB, Werner C, Taubenberger A. Precision Hydrogels for the Study of Cancer Cell Mechanobiology. Adv Healthc Mater 2023; 12:e2202514. [PMID: 36826799 PMCID: PMC11468035 DOI: 10.1002/adhm.202202514] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 01/23/2023] [Indexed: 02/25/2023]
Abstract
Cancer progression is associated with extensive remodeling of the tumor microenvironment (TME), resulting in alterations of biochemical and biophysical cues that affect both cancer and stromal cells. In particular, the mechanical characteristics of the TME extracellular matrix undergo significant changes. Bioengineered polymer hydrogels can be instrumental to systematically explore how mechanically changed microenvironments impact cancer cell behavior, including proliferation, survival, drug resistance, and invasion. This article reviews studies that have explored the impact of different mechanical cues of the cells' 3D microenvironment on cancer cell behavior using hydrogel-based in vitro models. In particular, advanced engineering strategies are highlighted for tailored hydrogel matrices recapitulating the TME's micrometer- and sub-micrometer-scale architectural and mechanical features, while accounting for its intrinsically heterogenic and dynamic nature. It is anticipated that such precision hydrogel systems will further the understanding of cancer mechanobiology.
Collapse
Affiliation(s)
- Jana Sievers
- Max Bergmann Center of Biomaterials DresdenLeibniz Institute for Polymer Research DresdenHohe Str. 601069DresdenGermany
| | - Vaibhav Mahajan
- Center for Molecular and Cellular Bioengineering (CMCB)BIOTECTU Dresden01307DresdenGermany
| | - Petra B. Welzel
- Max Bergmann Center of Biomaterials DresdenLeibniz Institute for Polymer Research DresdenHohe Str. 601069DresdenGermany
| | - Carsten Werner
- Max Bergmann Center of Biomaterials DresdenLeibniz Institute for Polymer Research DresdenHohe Str. 601069DresdenGermany
- Center of Regenerative Therapies Dresden and Cluster of Excellence Physics of LifeTU Dresden01062DresdenGermany
| | - Anna Taubenberger
- Max Bergmann Center of Biomaterials DresdenLeibniz Institute for Polymer Research DresdenHohe Str. 601069DresdenGermany
- Center for Molecular and Cellular Bioengineering (CMCB)BIOTECTU Dresden01307DresdenGermany
| |
Collapse
|
74
|
Ruscone M, Montagud A, Chavrier P, Destaing O, Bonnet I, Zinovyev A, Barillot E, Noël V, Calzone L. Multiscale model of the different modes of cancer cell invasion. Bioinformatics 2023; 39:btad374. [PMID: 37289551 PMCID: PMC10293590 DOI: 10.1093/bioinformatics/btad374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 05/25/2023] [Accepted: 06/07/2023] [Indexed: 06/10/2023] Open
Abstract
MOTIVATION Mathematical models of biological processes altered in cancer are built using the knowledge of complex networks of signaling pathways, detailing the molecular regulations inside different cell types, such as tumor cells, immune and other stromal cells. If these models mainly focus on intracellular information, they often omit a description of the spatial organization among cells and their interactions, and with the tumoral microenvironment. RESULTS We present here a model of tumor cell invasion simulated with PhysiBoSS, a multiscale framework, which combines agent-based modeling and continuous time Markov processes applied on Boolean network models. With this model, we aim to study the different modes of cell migration and to predict means to block it by considering not only spatial information obtained from the agent-based simulation but also intracellular regulation obtained from the Boolean model. Our multiscale model integrates the impact of gene mutations with the perturbation of the environmental conditions and allows the visualization of the results with 2D and 3D representations. The model successfully reproduces single and collective migration processes and is validated on published experiments on cell invasion. In silico experiments are suggested to search for possible targets that can block the more invasive tumoral phenotypes. AVAILABILITY AND IMPLEMENTATION https://github.com/sysbio-curie/Invasion_model_PhysiBoSS.
Collapse
Affiliation(s)
- Marco Ruscone
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
- Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | | | - Philippe Chavrier
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris, France
| | - Olivier Destaing
- Institute for Advanced Biosciences, Centre de Recherche Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, France
| | - Isabelle Bonnet
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, Paris, France
| | - Andrei Zinovyev
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
| | - Emmanuel Barillot
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
| | - Vincent Noël
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
| | - Laurence Calzone
- Institut Curie, Université PSL, F-75005 Paris, France
- INSERM, U900, F-75005 Paris, France
- Mines ParisTech, Université PSL, F-75005 Paris, France
| |
Collapse
|
75
|
Urciuolo A, Giobbe GG, Dong Y, Michielin F, Brandolino L, Magnussen M, Gagliano O, Selmin G, Scattolini V, Raffa P, Caccin P, Shibuya S, Scaglioni D, Wang X, Qu J, Nikolic M, Montagner M, Galea GL, Clevers H, Giomo M, De Coppi P, Elvassore N. Hydrogel-in-hydrogel live bioprinting for guidance and control of organoids and organotypic cultures. Nat Commun 2023; 14:3128. [PMID: 37253730 PMCID: PMC10229611 DOI: 10.1038/s41467-023-37953-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/06/2023] [Indexed: 06/01/2023] Open
Abstract
Three-dimensional hydrogel-based organ-like cultures can be applied to study development, regeneration, and disease in vitro. However, the control of engineered hydrogel composition, mechanical properties and geometrical constraints tends to be restricted to the initial time of fabrication. Modulation of hydrogel characteristics over time and according to culture evolution is often not possible. Here, we overcome these limitations by developing a hydrogel-in-hydrogel live bioprinting approach that enables the dynamic fabrication of instructive hydrogel elements within pre-existing hydrogel-based organ-like cultures. This can be achieved by crosslinking photosensitive hydrogels via two-photon absorption at any time during culture. We show that instructive hydrogels guide neural axon directionality in growing organotypic spinal cords, and that hydrogel geometry and mechanical properties control differential cell migration in developing cancer organoids. Finally, we show that hydrogel constraints promote cell polarity in liver organoids, guide small intestinal organoid morphogenesis and control lung tip bifurcation according to the hydrogel composition and shape.
Collapse
Affiliation(s)
- Anna Urciuolo
- Dept. of Molecular Medicine, University of Padova, Padova, Italy.
- Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy.
| | - Giovanni Giuseppe Giobbe
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Yixiao Dong
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China
| | - Federica Michielin
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Luca Brandolino
- Dept. of Industrial Engineering, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Michael Magnussen
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Onelia Gagliano
- Dept. of Industrial Engineering, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine, Padova, Italy
| | - Giulia Selmin
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | | | - Paolo Raffa
- Istituto di Ricerca Pediatrica, Città della Speranza, Padova, Italy
| | - Paola Caccin
- Dept. of Biomedical Science, University of Padova, Padova, Italy
| | - Soichi Shibuya
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Dominic Scaglioni
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Xuechun Wang
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China
| | - Ju Qu
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, China
| | - Marko Nikolic
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Marco Montagner
- Dept. of Molecular Medicine, University of Padova, Padova, Italy
| | - Gabriel L Galea
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
| | - Hans Clevers
- Hubrecht Institute, KNAW and University Medical Center, Utrecht, The Netherlands
- Pharma Research and Early Development (pRED) of Roche, Basel, Switzerland
| | - Monica Giomo
- Dept. of Industrial Engineering, University of Padova, Padova, Italy
| | - Paolo De Coppi
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK
- Dept. of Specialist Neonatal and Paediatric Surgery, Great Ormond Street Hospital, London, UK
| | - Nicola Elvassore
- GOSICH Zayed Centre for Research into Rare Disease in Children, University College London, London, UK.
- Dept. of Industrial Engineering, University of Padova, Padova, Italy.
- Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
76
|
Almagro J, Messal HA. Volume imaging to interrogate cancer cell-tumor microenvironment interactions in space and time. Front Immunol 2023; 14:1176594. [PMID: 37261345 PMCID: PMC10228654 DOI: 10.3389/fimmu.2023.1176594] [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: 02/28/2023] [Accepted: 04/26/2023] [Indexed: 06/02/2023] Open
Abstract
Volume imaging visualizes the three-dimensional (3D) complexity of tumors to unravel the dynamic crosstalk between cancer cells and the heterogeneous landscape of the tumor microenvironment (TME). Tissue clearing and intravital microscopy (IVM) constitute rapidly progressing technologies to study the architectural context of such interactions. Tissue clearing enables high-resolution imaging of large samples, allowing for the characterization of entire tumors and even organs and organisms with tumors. With IVM, the dynamic engagement between cancer cells and the TME can be visualized in 3D over time, allowing for acquisition of 4D data. Together, tissue clearing and IVM have been critical in the examination of cancer-TME interactions and have drastically advanced our knowledge in fundamental cancer research and clinical oncology. This review provides an overview of the current technical repertoire of fluorescence volume imaging technologies to study cancer and the TME, and discusses how their recent applications have been utilized to advance our fundamental understanding of tumor architecture, stromal and immune infiltration, vascularization and innervation, and to explore avenues for immunotherapy and optimized chemotherapy delivery.
Collapse
Affiliation(s)
- Jorge Almagro
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, United States
| | - Hendrik A. Messal
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan, Amsterdam, Netherlands
| |
Collapse
|
77
|
Almeida JA, Mathur J, Lee YL, Sarker B, Pathak A. Mechanically primed cells transfer memory to fibrous matrices for invasion across environments of distinct stiffness and dimensionality. Mol Biol Cell 2023; 34:ar54. [PMID: 36696158 PMCID: PMC10208097 DOI: 10.1091/mbc.e22-10-0469] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
Cells sense and migrate across mechanically dissimilar environments throughout development and disease progression. However, it remains unclear whether mechanical memory of past environments empowers cells to navigate new, three-dimensional extracellular matrices. Here, we show that cells previously primed on stiff, compared with soft, matrices generate a higher level of forces to remodel collagen fibers and promote invasion. This priming advantage persists in dense or stiffened collagen. We explain this memory-dependent, cross-environment cell invasion through a lattice-based model wherein stiff-primed cellular forces remodel collagen and minimize energy required for future cell invasion. According to our model, cells transfer their mechanical memory to the matrix via collagen alignment and tension, and this remodeled matrix informs future cell invasion. Thus, memory-laden cells overcome mechanosensing of softer or challenging future environments via a cell-matrix transfer of memory. Consistent with model predictions, depletion of yes-associated protein destabilizes the cellular memory required for collagen remodeling before invasion. We release tension in collagen fibers via laser ablation and disable fiber remodeling by lysyl-oxidase inhibition, both of which disrupt cell-to-matrix transfer of memory and hamper cross-environment invasion. These results have implications for cancer, fibrosis, and aging, where a potential cell-to-matrix transfer of mechanical memory of cells may generate a prolonged cellular response.
Collapse
Affiliation(s)
- José A. Almeida
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Jairaj Mathur
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| | - Ye Lim Lee
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
| | - Bapi Sarker
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130
- Department of Mechanical Engineering & Materials Science, Washington University, St. Louis, MO 63130
| |
Collapse
|
78
|
Dupin I, Eyraud E, Maurat É, Sac-Épée JM, Vallois P. Probabilistic cellular automata modelling of intercellular interactions in airways: complex pattern formation in patients with chronic obstructive pulmonary disease. J Theor Biol 2023; 564:111448. [PMID: 36878400 DOI: 10.1016/j.jtbi.2023.111448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 02/16/2023] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
Chronic obstructive pulmonary disease (COPD) is a highly prevalent lung disease characterized by chronic inflammation and tissue remodeling possibly induced by unusual interactions between fibrocytes and CD8+ T lymphocytes in the peribronchial area. To investigate this phenomenon, we developed a probabilistic cellular automata type model where the two types of cells follow simple local interaction rules taking into account cell death, proliferation, migration and infiltration. We conducted a rigorous mathematical analysis using multiscale experimental data obtained in control and disease conditions to estimate the model's parameters accurately. The simulation of the model is straightforward to implement, and two distinct patterns emerged that we can analyse quantitatively. In particular, we show that the change in fibrocyte density in the COPD condition is mainly the consequence of their infiltration into the lung during exacerbations, suggesting possible explanations for experimental observations in normal and COPD tissue. Our integrated approach that combines a probabilistic cellular automata model and experimental findings will provide further insights into COPD in future studies.
Collapse
Affiliation(s)
- Isabelle Dupin
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France.
| | - Edmée Eyraud
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France
| | - Élise Maurat
- Univ-Bordeaux, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France; INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, F-33000 Bordeaux, France
| | | | - Pierre Vallois
- Université de Lorraine, CNRS, Inria, IECL., F-54000 Nancy, France
| |
Collapse
|
79
|
Stöberl S, Balles M, Kellerer T, Rädler JO. Photolithographic microfabrication of hydrogel clefts for cell invasion studies. LAB ON A CHIP 2023; 23:1886-1895. [PMID: 36867426 DOI: 10.1039/d2lc01105k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Invasion of migrating cells into surrounding tissue plays a key role in cancer metastasis and immune response. In order to assess invasiveness, most in vitro invasion assays measure the degree to which cells migrate between microchambers that provide a chemoattractant gradient across a polymeric membrane with defined pores. However, in real tissue cells experience soft, mechanically deformable microenvironments. Here we introduce RGD-functionalized hydrogel structures that present pressurized clefts for invasive migration of cells between reservoirs maintaining a chemotactic gradient. Using UV-photolithography, equally spaced blocks of polyethylene glycol-norbornene (PEG-NB) hydrogels are formed, which subsequently swell and close the interjacent gaps. The swelling ratio and final contours of the hydrogel blocks were determined using confocal microscopy confirming a swelling induced closure of the structures. The velocity profile of cancer cells transmigrating through the clefts, which we name 'sponge clamp', is found to depend on the elastic modulus as well as the gap size between the swollen blocks. The 'sponge clamp' discriminates the invasiveness of two distinct cell lines, MDA-MB-231 and HT-1080. The approach provides soft 3D-microstructures mimicking invasion conditions in extracellular matrix.
Collapse
Affiliation(s)
- Stefan Stöberl
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
| | - Miriam Balles
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
| | - Thomas Kellerer
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
- Department of Applied Science and Mechatronics, University of Applied Science, Munich, Germany
| | - Joachim O Rädler
- Faculty of Physics and Center for NanoScience, Ludwig Maximilians-University, Munich, Germany.
| |
Collapse
|
80
|
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: 1.0] [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
|
81
|
Yamamoto A, Doak AE, Cheung KJ. Orchestration of Collective Migration and Metastasis by Tumor Cell Clusters. ANNUAL REVIEW OF PATHOLOGY 2023; 18:231-256. [PMID: 36207009 DOI: 10.1146/annurev-pathmechdis-031521-023557] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Metastatic dissemination has lethal consequences for cancer patients. Accruing evidence supports the hypothesis that tumor cells can migrate and metastasize as clusters of cells while maintaining contacts with one another. Collective metastasis enables tumor cells to colonize secondary sites more efficiently, resist cell death, and evade the immune system. On the other hand, tumor cell clusters face unique challenges for dissemination particularly during systemic dissemination. Here, we review recent progress toward understanding how tumor cell clusters overcome these disadvantages as well as mechanisms they utilize to gain advantages throughout the metastatic process. We consider useful models for studying collective metastasis and reflect on how the study of collective metastasis suggests new opportunities for eradicating and preventing metastatic disease.
Collapse
Affiliation(s)
- Ami Yamamoto
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA; , , .,Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Andrea E Doak
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA; , , .,Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington, USA
| | - Kevin J Cheung
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Center, Seattle, Washington, USA; , ,
| |
Collapse
|
82
|
Smits D, Khalil AA. Multimodal Techniques to Study Tumor Growth, Basement Membrane Breaching, and Invasion in 3D Matrices. Methods Mol Biol 2023; 2608:281-303. [PMID: 36653714 DOI: 10.1007/978-1-0716-2887-4_17] [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: 01/20/2023]
Abstract
Cancer-derived organoids and three-dimensional (3D) extracellular matrix (ECM) are taking center stage as in vitro models to study neoplastic cell behavior, since they recapitulate the heterogeneous cellular composition of tumors and their extracellular environment. In combination with imaging and molecular/biochemical techniques, 3D organoid models have contributed substantially to our knowledge about the cellular and molecular mechanisms that regulate the growth of tumors and invasion into the surrounding tissue. We here outline a set of protocols that describe culturing of cancer-derived organoids in 3D matrices and various strategies that allow modeling of tumor growth, tumor cell penetration into basement membranes, and invasion into Collagen I-rich ECM. Furthermore, we specify protocols for subsequent handling of organoids cultured in 3D ECM for confocal microscopy and analysis of gene expression at the protein and mRNA level. Although we here use breast cancer-derived organoids, these protocols can be directly applied or adapted for organoids derived from other cancer types or healthy tissues. Thus, in addition to investigating cell behavior of multiple cancer types, the combination of protocols described here may be used to study processes such as cell differentiation and migration during homeostasis and normal development.
Collapse
Affiliation(s)
- Daan Smits
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Cell Biology, Radboudumc, Nijmegen, The Netherlands
| | - Antoine A Khalil
- Center for Molecular Medicine (CMM), University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
83
|
Law RA, Kiepas A, Desta HE, Perez Ipiña E, Parlani M, Lee SJ, Yankaskas CL, Zhao R, Mistriotis P, Wang N, Gu Z, Kalab P, Friedl P, Camley BA, Konstantopoulos K. Cytokinesis machinery promotes cell dissociation from collectively migrating strands in confinement. SCIENCE ADVANCES 2023; 9:eabq6480. [PMID: 36630496 PMCID: PMC9833664 DOI: 10.1126/sciadv.abq6480] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Cells tune adherens junction dynamics to regulate epithelial integrity in diverse (patho)physiological processes, including cancer metastasis. We hypothesized that the spatially confining architecture of peritumor stroma promotes metastatic cell dissemination by remodeling cell-cell adhesive interactions. By combining microfluidics with live-cell imaging, FLIM/FRET biosensors, and optogenetic tools, we show that confinement induces leader cell dissociation from cohesive ensembles. Cell dissociation is triggered by myosin IIA (MIIA) dismantling of E-cadherin cell-cell junctions, as recapitulated by a mathematical model. Elevated MIIA contractility is controlled by RhoA/ROCK activation, which requires distinct guanine nucleotide exchange factors (GEFs). Confinement activates RhoA via nucleocytoplasmic shuttling of the cytokinesis-regulatory proteins RacGAP1 and Ect2 and increased microtubule dynamics, which results in the release of active GEF-H1. Thus, confining microenvironments are sufficient to induce cell dissemination from primary tumors by remodeling E-cadherin cell junctions via the interplay of microtubules, nuclear trafficking, and RhoA/ROCK/MIIA pathway and not by down-regulating E-cadherin expression.
Collapse
Affiliation(s)
- 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
| | - Alexander Kiepas
- 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
| | - Habben E. Desta
- 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
| | - Emiliano Perez Ipiña
- William H. Miller III Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Maria Parlani
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Se Jong Lee
- 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
| | - Christopher L. Yankaskas
- 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
| | - 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
| | - 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
| | - Nianchao Wang
- 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
| | - Zhizhan Gu
- 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
- Corresponding author. (K.K.); (Z.G.)
| | - Petr Kalab
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, 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 Center, 3584 Utrecht, Netherlands
| | - Brian A. Camley
- William H. Miller III Department of Physics and Astronomy, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - 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
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Department of Oncology, Johns Hopkins University, Baltimore, MD 21205, USA
- Corresponding author. (K.K.); (Z.G.)
| |
Collapse
|
84
|
Chan TJ, Zhang X, Mak M. Biophysical informatics reveals distinctive phenotypic signatures and functional diversity of single-cell lineages. Bioinformatics 2023; 39:6969104. [PMID: 36610710 PMCID: PMC9825265 DOI: 10.1093/bioinformatics/btac833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/11/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022] Open
Abstract
MOTIVATION In this work, we present an analytical method for quantifying both single-cell morphologies and cell network topologies of tumor cell populations and use it to predict 3D cell behavior. RESULTS We utilized a supervised deep learning approach to perform instance segmentation on label-free live cell images across a wide range of cell densities. We measured cell shape properties and characterized network topologies for 136 single-cell clones derived from the YUMM1.7 and YUMMER1.7 mouse melanoma cell lines. Using an unsupervised clustering algorithm, we identified six distinct morphological subclasses. We further observed differences in tumor growth and invasion dynamics across subclasses in an in vitro 3D spheroid model. Compared to existing methods for quantifying 2D or 3D phenotype, our analytical method requires less time, needs no specialized equipment and is capable of much higher throughput, making it ideal for applications such as high-throughput drug screening and clinical diagnosis. AVAILABILITY AND IMPLEMENTATION https://github.com/trevor-chan/Melanoma_NetworkMorphology. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Trevor J Chan
- Department of Bioengineering, Yale University, New Haven, CT 06511, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xingjian Zhang
- Department of Bioengineering, Yale University, New Haven, CT 06511, USA
| | | |
Collapse
|
85
|
Sadhukhan S, Nandi SK. On the origin of universal cell shape variability in confluent epithelial monolayers. eLife 2022; 11:e76406. [PMID: 36563034 PMCID: PMC9833828 DOI: 10.7554/elife.76406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 12/22/2022] [Indexed: 12/24/2022] Open
Abstract
Cell shape is fundamental in biology. The average cell shape can influence crucial biological functions, such as cell fate and division orientation. But cell-to-cell shape variability is often regarded as noise. In contrast, recent works reveal that shape variability in diverse epithelial monolayers follows a nearly universal distribution. However, the origin and implications of this universality remain unclear. Here, assuming contractility and adhesion are crucial for cell shape, characterized via aspect ratio (r), we develop a mean-field analytical theory for shape variability. We find that all the system-specific details combine into a single parameter α that governs the probability distribution function (PDF) of r; this leads to a universal relation between the standard deviation and the average of r. The PDF for the scaled r is not strictly but nearly universal. In addition, we obtain the scaled area distribution, described by the parameter μ. Information of α and μ together can distinguish the effects of changing physical conditions, such as maturation, on different system properties. We have verified the theory via simulations of two distinct models of epithelial monolayers and with existing experiments on diverse systems. We demonstrate that in a confluent monolayer, average shape determines both the shape variability and dynamics. Our results imply that cell shape distribution is inevitable, where a single parameter describes both statics and dynamics and provides a framework to analyze and compare diverse epithelial systems. In contrast to existing theories, our work shows that the universal properties are consequences of a mathematical property and should be valid in general, even in the fluid regime.
Collapse
|
86
|
Hohmann U, von Widdern JC, Ghadban C, Giudice MCL, Lemahieu G, Cavalcanti-Adam EA, Dehghani F, Hohmann T. Jamming Transitions in Astrocytes and Glioblastoma Are Induced by Cell Density and Tension. Cells 2022; 12:cells12010029. [PMID: 36611824 PMCID: PMC9818602 DOI: 10.3390/cells12010029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Collective behavior of cells emerges from coordination of cell-cell-interactions and is important to wound healing, embryonic and tumor development. Depending on cell density and cell-cell interactions, a transition from a migratory, fluid-like unjammed state to a more static and solid-like jammed state or vice versa can occur. Here, we analyze collective migration dynamics of astrocytes and glioblastoma cells using live cell imaging. Furthermore, atomic force microscopy, traction force microscopy and spheroid generation assays were used to study cell adhesion, traction and mechanics. Perturbations of traction and adhesion were induced via ROCK or myosin II inhibition. Whereas astrocytes resided within a non-migratory, jammed state, glioblastoma were migratory and unjammed. Furthermore, we demonstrated that a switch from an unjammed to a jammed state was induced upon alteration of the equilibrium between cell-cell-adhesion and tension from adhesion to tension dominated, via inhibition of ROCK or myosin II. Such behavior has implications for understanding the infiltration of the brain by glioblastoma cells and may help to identify new strategies to develop anti-migratory drugs and strategies for glioblastoma-treatment.
Collapse
Affiliation(s)
- Urszula Hohmann
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Julian Cardinal von Widdern
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Chalid Ghadban
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Maria Cristina Lo Giudice
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Grégoire Lemahieu
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | | | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Tim Hohmann
- Department of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
- Correspondence:
| |
Collapse
|
87
|
La Porta CAM, Zapperi S. Artificial intelligence in breast cancer diagnostics. Cell Rep Med 2022; 3:100851. [PMID: 36543102 PMCID: PMC9798018 DOI: 10.1016/j.xcrm.2022.100851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 12/24/2022]
Abstract
Since breast cancer deaths are mainly due to metastasis, predicting the risk that a primary tumor will develop metastasis after a first diagnosis is a central issue that could be addressed by artificial intelligence. To overcome the problem posed by limited availability of standardized datasets, algorithms should include biological insight.
Collapse
Affiliation(s)
- Caterina AM. La Porta
- Department of Environmental Science and Policy, Center for Complexity & Biosystems, University of Milan, via Celoria 10, 20133 Milan, Italy,CNR - Consiglio Nazionale delle Ricerche, Istituto di Biofisica, via Celoria 10, 20133 Milan, Italy,Corresponding author
| | - Stefano Zapperi
- Department of Physics, Center for Complexity & Biosystems, University of Milan, via Celoria 16, 20133 Milan, Italy,CNR - Consiglio Nazionale delle Ricerche, Istituto di Chimica della Materia Condensata e di Tecnologie per l'Energia, Via R. Cozzi 53, 20125 Milano, Italy
| |
Collapse
|
88
|
Perrin L, Belova E, Bayarmagnai B, Tüzel E, Gligorijevic B. Invadopodia enable cooperative invasion and metastasis of breast cancer cells. Commun Biol 2022; 5:758. [PMID: 35915226 PMCID: PMC9343607 DOI: 10.1038/s42003-022-03642-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Invasive and non-invasive cancer cells can invade together during cooperative invasion. However, the events leading to it, role of the epithelial-mesenchymal transition and the consequences this may have on metastasis are unknown. In this study, we demonstrate that the isogenic 4T1 and 67NR breast cancer cells sort from each other in 3D spheroids, followed by cooperative invasion. By time-lapse microscopy, we show that the invasive 4T1 cells move more persistently compared to non-invasive 67NR, sorting and accumulating at the spheroid-matrix interface, a process dependent on cell-matrix adhesions and independent from E-cadherin cell-cell adhesions. Elimination of invadopodia in 4T1 cells blocks invasion, demonstrating that invadopodia requirement is limited to leader cells. Importantly, we demonstrate that cells with and without invadopodia can also engage in cooperative metastasis in preclinical mouse models. Altogether, our results suggest that a small number of cells with invadopodia can drive the metastasis of heterogeneous cell clusters. Cooperative invasion requires the formation of invadopodia in the leader cells, and a small number of leader cells may be enough to facilitate cooperative invasion and metastasis, including non-invadopodia forming cancer cells.
Collapse
|
89
|
Franchi M, Karamanos KA, Cappadone C, Calonghi N, Greco N, Franchi L, Onisto M, Masola V. Substrate Type and Concentration Differently Affect Colon Cancer Cells Ultrastructural Morphology, EMT Markers, and Matrix Degrading Enzymes. Biomolecules 2022; 12:1786. [PMID: 36551219 PMCID: PMC9775446 DOI: 10.3390/biom12121786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Aim of the study was to understand the behavior of colon cancer LoVo-R cells (doxorubicin-resistant) vs. LoVo-S (doxorubicin sensitive) in the initial steps of extracellular matrix (ECM) invasion. We investigated how the matrix substrates Matrigel and type I collagen-mimicking the basement membrane (BM) and the normal or desmoplastic lamina propria, respectively-could affect the expression of epithelial-to-mesenchymal transition (EMT) markers, matrix-degrading enzymes, and phenotypes. Gene expression with RT-qPCR, E-cadherin protein expression using Western blot, and phenotypes using scanning electron microscopy (SEM) were analyzed. The type and different concentrations of matrix substrates differently affected colon cancer cells. In LoVo-S cells, the higher concentrated collagen, mimicking the desmoplastic lamina propria, strongly induced EMT, as also confirmed by the expression of Snail, metalloproteases (MMPs)-2, -9, -14 and heparanase (HPSE), as well as mesenchymal phenotypes. Stimulation in E-cadherin expression in LoVo-S groups suggests that these cells develop a hybrid EMT phenotype. Differently, LoVo-R cells did not increase their aggressiveness: no changes in EMT markers, matrix effectors, and phenotypes were evident. The low influence of ECM components in LoVo-R cells might be related to their intrinsic aggressiveness related to chemoresistance. These results improve understanding of the critical role of tumor microenvironment in colon cancer cell invasion, driving the development of new therapeutic approaches.
Collapse
Affiliation(s)
- Marco Franchi
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy
| | | | - Concettina Cappadone
- Department of Pharmacy and Biotechnologies, University of Bologna, 40126 Bologna, Italy
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnologies, University of Bologna, 40126 Bologna, Italy
| | - Nicola Greco
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Leonardo Franchi
- Department of Medicine, University of Bologna, 40126 Bologna, Italy
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Valentina Masola
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
90
|
Daulagala AC, Kourtidis A. ECM Substrates Impact RNAi Localization at Adherens Junctions of Colon Epithelial Cells. Cells 2022; 11:3740. [PMID: 36497003 PMCID: PMC9737857 DOI: 10.3390/cells11233740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/08/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
The extracellular matrix (ECM) plays crucial roles in tissue homeostasis. Abnormalities in ECM composition are associated with pathological conditions, such as fibrosis and cancer. These ECM alterations are sensed by the epithelium and can influence its behavior through crosstalk with other mechanosensitive complexes, including the adherens junctions (AJs). We have previously shown that the AJs, through their component PLEKHA7, recruit the RNAi machinery to regulate miRNA levels and function. We have particularly shown that the junctional localization of RNAi components is critical for their function. Here, we investigated whether different ECM substrates can influence the junctional localization of RNAi complexes. To do this, we plated colon epithelial Caco2 cells on four key ECM substrates found in the colon under normal or pathogenic conditions, namely laminin, fibronectin, collagen I, and collagen IV, and we examined the subcellular distribution of PLEKHA7, and of the key RNAi components AGO2 and DROSHA. Fibronectin and collagen I negatively impacted the junctional localization of PLEKHA7, AGO2, and DROSHA when compared to laminin. Furthermore, fibronectin, collagen I, and collagen IV disrupted interactions of AGO2 and DROSHA with their essential partners GW182 and DGCR8, respectively, both at AJs and throughout the cell. Combinations of all substrates with fibronectin also negatively impacted junctional localization of PLEKHA7 and AGO2. Additionally, collagen I triggered accumulation of DROSHA at tri-cellular junctions, while both collagen I and collagen IV resulted in DROSHA accumulation at basal areas of cell-cell contact. Altogether, fibronectin and collagens I and IV, which are elevated in the stroma of fibrotic and cancerous tissues, altered localization patterns and disrupted complex formation of PLEKHA7 and RNAi components. Combined with our prior studies showing that apical junctional localization of the PLEKHA7-RNAi complex is critical for regulating tumor-suppressing miRNAs, this work points to a yet unstudied mechanism that could contribute to epithelial cell transformation.
Collapse
Affiliation(s)
| | - Antonis Kourtidis
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
91
|
Perrin L, Gligorijevic B. Proteolytic and mechanical remodeling of the extracellular matrix by invadopodia in cancer. Phys Biol 2022; 20:10.1088/1478-3975/aca0d8. [PMID: 36343366 PMCID: PMC9942491 DOI: 10.1088/1478-3975/aca0d8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022]
Abstract
Cancer invasion and metastasis require remodeling of the adjacent extracellular matrix (ECM). In this mini review, we will cover the mechanisms of proteolytic degradation and the mechanical remodeling of the ECM by cancer cells, with a focus on invadopodia. Invadopodia are membrane protrusions unique to cancer cells, characterized by an actin core and by the focal degradation of ECM via matrix metalloproteases (MMPs). While ECM can also be remodeled, at lower levels, by focal adhesions, or internal collagen digestion, invadopodia are now recognized as the major mechanism for MMP-dependent pericellular ECM degradation by cancer cells. Recent evidence suggests that the completion of epithelial-mesenchymal transition may be dispensable for invadopodia and metastasis, and that invadopodia are required not only for mesenchymal, single cell invasion, but also for collective invasion. During collective invasion, invadopodia was then shown to be located in leader cells, allowing follower cells to move via cooperation. Collectively, this suggests that invadopodia function may be a requirement not only for later steps of metastasis, but also for early invasion of epithelial cells into the stromal tissue. Over the last decade, invadopodia studies have transitioned into in 3D andin vivosettings, leading to the confirmation of their essential role in metastasis in preclinical animal models. In summary, invadopodia may hold a great potential for individual risk assessment as a prognostic marker for metastasis, as well as a therapeutic target.
Collapse
Affiliation(s)
- L. Perrin
- Bioengineering Department, Temple University, Philadelphia PA, USA
- Present address, Institut Curie, Paris, France
| | - B. Gligorijevic
- Bioengineering Department, Temple University, Philadelphia PA, USA
- Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia PA, USA
| |
Collapse
|
92
|
Scheele CLGJ, Herrmann D, Yamashita E, Celso CL, Jenne CN, Oktay MH, Entenberg D, Friedl P, Weigert R, Meijboom FLB, Ishii M, Timpson P, van Rheenen J. Multiphoton intravital microscopy of rodents. NATURE REVIEWS. METHODS PRIMERS 2022; 2:89. [PMID: 37621948 PMCID: PMC10449057 DOI: 10.1038/s43586-022-00168-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/12/2022] [Indexed: 08/26/2023]
Abstract
Tissues are heterogeneous with respect to cellular and non-cellular components and in the dynamic interactions between these elements. To study the behaviour and fate of individual cells in these complex tissues, intravital microscopy (IVM) techniques such as multiphoton microscopy have been developed to visualize intact and live tissues at cellular and subcellular resolution. IVM experiments have revealed unique insights into the dynamic interplay between different cell types and their local environment, and how this drives morphogenesis and homeostasis of tissues, inflammation and immune responses, and the development of various diseases. This Primer introduces researchers to IVM technologies, with a focus on multiphoton microscopy of rodents, and discusses challenges, solutions and practical tips on how to perform IVM. To illustrate the unique potential of IVM, several examples of results are highlighted. Finally, we discuss data reproducibility and how to handle big imaging data sets.
Collapse
Affiliation(s)
- Colinda L. G. J. Scheele
- Laboratory for Intravital Imaging and Dynamics of Tumor Progression, VIB Center for Cancer Biology, KU Leuven, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - David Herrmann
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Department, Sydney, New South Wales, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Erika Yamashita
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Cristina Lo Celso
- Department of Life Sciences and Centre for Hematology, Imperial College London, London, UK
- Sir Francis Crick Institute, London, UK
| | - Craig N. Jenne
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Maja H. Oktay
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - David Entenberg
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
- Integrated Imaging Program, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY, USA
| | - Peter Friedl
- Department of Cell Biology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, Netherlands
- David H. Koch Center for Applied Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roberto Weigert
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Franck L. B. Meijboom
- Department of Population Health Sciences, Sustainable Animal Stewardship, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- Faculty of Humanities, Ethics Institute, Utrecht University, Utrecht, Netherlands
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
- WPI-Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory of Bioimaging and Drug Discovery, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Paul Timpson
- Cancer Ecosystems Program, Garvan Institute of Medical Research and The Kinghorn Cancer Centre, Cancer Department, Sydney, New South Wales, Australia
- St. Vincent’s Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
93
|
Ma J, Zhang L, Li S, Liu H. BRPCA: Bounded Robust Principal Component Analysis to Incorporate Similarity Network for N7-Methylguanosine(m 7G) Site-Disease Association Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:3295-3306. [PMID: 34469307 DOI: 10.1109/tcbb.2021.3109055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Recent studies have revealed that N7-methylguanosine(m7G) plays a pivotal role in various biological processes and disease pathogenesis. To date, transcriptome-wide m7G modification sites have been identified by high-throughput sequencing approaches, and some related information has been recorded in a few biological databases. However, the mechanism of site action in disease remains uncharted. Wet experiments can help identify true m7G sites with high confidence, but it is time-consuming to find the true ones in such a large number of sites, which will also cost too much. Thus, computational methods are emergently needed to predict the associations between m7G sites and various diseases, thus help to uncover potential active sites for specific diseases. In this article, we proposed a bounded robust principal component analysis (BRPCA) method to predict unknown m7G-disease association based on similarity information. Importantly, BRPCA tolerates the noise and redundancy existing in association and similarity information. Moreover, a suitable bounded constraint is incorporated into BRPCA to ensure that the predicted association scores locate in a meaningful interval. The extensive experiments demonstrate the superiority and robustness of the BRPCA.
Collapse
|
94
|
Cai G, Nguyen A, Bashirzadeh Y, Lin SS, Bi D, Liu AP. Compressive stress drives adhesion-dependent unjamming transitions in breast cancer cell migration. Front Cell Dev Biol 2022; 10:933042. [PMID: 36268514 PMCID: PMC9577106 DOI: 10.3389/fcell.2022.933042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular unjamming is the collective fluidization of cell motion and has been linked to many biological processes, including development, wound repair, and tumor growth. In tumor growth, the uncontrolled proliferation of cancer cells in a confined space generates mechanical compressive stress. However, because multiple cellular and molecular mechanisms may be operating simultaneously, the role of compressive stress in unjamming transitions during cancer progression remains unknown. Here, we investigate which mechanism dominates in a dense, mechanically stressed monolayer. We find that long-term mechanical compression triggers cell arrest in benign epithelial cells and enhances cancer cell migration in transitions correlated with cell shape, leading us to examine the contributions of cell–cell adhesion and substrate traction in unjamming transitions. We show that cadherin-mediated cell–cell adhesion regulates differential cellular responses to compressive stress and is an important driver of unjamming in stressed monolayers. Importantly, compressive stress does not induce the epithelial–mesenchymal transition in unjammed cells. Furthermore, traction force microscopy reveals the attenuation of traction stresses in compressed cells within the bulk monolayer regardless of cell type and motility. As traction within the bulk monolayer decreases with compressive pressure, cancer cells at the leading edge of the cell layer exhibit sustained traction under compression. Together, strengthened intercellular adhesion and attenuation of traction forces within the bulk cell sheet under compression lead to fluidization of the cell layer and may impact collective cell motion in tumor development and breast cancer progression.
Collapse
Affiliation(s)
- Grace Cai
- Applied Physics Program, University of Michigan, Ann Arbor, MI, United States
| | - Anh Nguyen
- Department of Physics, Northeastern University, Boston, MA, United States
| | - Yashar Bashirzadeh
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Shan-Shan Lin
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Dapeng Bi
- Department of Physics, Northeastern University, Boston, MA, United States
| | - Allen P. Liu
- Applied Physics Program, University of Michigan, Ann Arbor, MI, United States
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
- Department of Biophysics, University of Michigan, Ann Arbor, MI, United States
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Allen P. Liu,
| |
Collapse
|
95
|
Yamada KM, Doyle AD, Lu J. Cell-3D matrix interactions: recent advances and opportunities. Trends Cell Biol 2022; 32:883-895. [PMID: 35410820 PMCID: PMC9464680 DOI: 10.1016/j.tcb.2022.03.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/03/2023]
Abstract
Tissues consist of cells and their surrounding extracellular matrix (ECM). Cell-ECM interactions play crucial roles in embryonic development, differentiation, tissue remodeling, and diseases including fibrosis and cancer. Recent research advances in characterizing cell-matrix interactions include detailed descriptions of hundreds of ECM and associated molecules, their complex intermolecular interactions in development and disease, identification of distinctive modes of cell migration in different 3D ECMs, and new insights into mechanisms of organ formation. Exploring the roles of the physical features of different ECM microenvironments and the bidirectional regulation of cell signaling and matrix organization emphasize the dynamic nature of these interactions, which can include feedback loops that exacerbate disease. Understanding mechanisms of cell-matrix interactions can potentially lead to targeted therapeutic interventions.
Collapse
Affiliation(s)
- Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Andrew D Doyle
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jiaoyang Lu
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
96
|
Guan X, Huang S. Advances in the application of 3D tumor models in precision oncology and drug screening. Front Bioeng Biotechnol 2022; 10:1021966. [PMID: 36246388 PMCID: PMC9555934 DOI: 10.3389/fbioe.2022.1021966] [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: 08/18/2022] [Accepted: 09/13/2022] [Indexed: 11/29/2022] Open
Abstract
Traditional tumor models cannot perfectly simulate the real state of tumors in vivo, resulting in the termination of many clinical trials. 3D tumor models’ technology provides new in vitro models that bridge the gap between in vitro and in vivo findings, and organoids maintain the properties of the original tissue over a long period of culture, which enables extensive research in this area. In addition, they can be used as a substitute for animal and in vitro models, and organoids can be established from patients’ normal and malignant tissues, with unique advantages in clinical drug development and in guiding individualized therapies. 3D tumor models also provide a promising platform for high-throughput research, drug and toxicity testing, disease modeling, and regenerative medicine. This report summarizes the 3D tumor model, including evidence regarding the 3D tumor cell culture model, 3D tumor slice model, and organoid culture model. In addition, it provides evidence regarding the application of 3D tumor organoid models in precision oncology and drug screening. The aim of this report is to elucidate the value of 3D tumor models in cancer research and provide a preclinical reference for the precise treatment of cancer patients.
Collapse
Affiliation(s)
- Xiaoyong Guan
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi, China
| | - Shigao Huang
- Department of Radiation Oncology, The First Affiliated Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Shigao Huang,
| |
Collapse
|
97
|
Sala F, Ficorella C, Osellame R, Käs JA, Martínez Vázquez R. Microfluidic Lab-on-a-Chip for Studies of Cell Migration under Spatial Confinement. BIOSENSORS 2022; 12:bios12080604. [PMID: 36004998 PMCID: PMC9405557 DOI: 10.3390/bios12080604] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022]
Abstract
Understanding cell migration is a key step in unraveling many physiological phenomena and predicting several pathologies, such as cancer metastasis. In particular, confinement has been proven to be a key factor in the cellular migration strategy choice. As our insight in the field improves, new tools are needed in order to empower biologists’ analysis capabilities. In this framework, microfluidic devices have been used to engineer the mechanical and spatial stimuli and to investigate cellular migration response in a more controlled way. In this work, we will review the existing technologies employed in the realization of microfluidic cellular migration assays, namely the soft lithography of PDMS and hydrogels and femtosecond laser micromachining. We will give an overview of the state of the art of these devices, focusing on the different geometrical configurations that have been exploited to study specific aspects of cellular migration. Our scope is to highlight the advantages and possibilities given by each approach and to envisage the future developments in in vitro migration studies under spatial confinement in microfluidic devices.
Collapse
Affiliation(s)
- Federico Sala
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Carlotta Ficorella
- Peter Debye Institute for Soft Matter Physics, University of Leipzig, 04109 Leipzig, Germany
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Josef A. Käs
- Peter Debye Institute for Soft Matter Physics, University of Leipzig, 04109 Leipzig, Germany
| | - Rebeca Martínez Vázquez
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
- Correspondence:
| |
Collapse
|
98
|
Tao J, Zhu L, Yakoub M, Reißfelder C, Loges S, Schölch S. Cell-Cell Interactions Drive Metastasis of Circulating Tumor Microemboli. Cancer Res 2022; 82:2661-2671. [PMID: 35856896 DOI: 10.1158/0008-5472.can-22-0906] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/27/2022] [Accepted: 06/13/2022] [Indexed: 11/16/2022]
Abstract
Circulating tumor cells are the cellular mediators of distant metastasis in solid malignancies. Their metastatic potential can be augmented by clustering with other tumor cells or nonmalignant cells, forming circulating tumor microemboli (CTM). Cell-cell interactions are key regulators within CTM that convey enhanced metastatic properties, including improved cell survival, immune evasion, and effective extravasation into distant organs. However, the cellular and molecular mechanism of CTM formation, as well as the biology of interactions between tumor cells and immune cells, platelets, and stromal cells in the circulation, remains to be determined. Here, we review the current literature on cell-cell interactions in homotypic and heterotypic CTM and provide perspectives on therapeutic strategies to attenuate CTM-mediated metastasis by targeting cell-cell interactions.
Collapse
Affiliation(s)
- Jianxin Tao
- JCCU Translational Surgical Oncology (A430), German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.,Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lei Zhu
- JCCU Translational Surgical Oncology (A430), German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.,Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Mina Yakoub
- JCCU Translational Surgical Oncology (A430), German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.,Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Reißfelder
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.,Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sonja Loges
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.,Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Personalized Oncology, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Sebastian Schölch
- JCCU Translational Surgical Oncology (A430), German Cancer Research Center (DKFZ), Heidelberg, Germany.,DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.,Department of Surgery, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
99
|
Pajic-Lijakovic I, Milivojevic M, Clark AG. Collective Cell Migration on Collagen-I Networks: The Impact of Matrix Viscoelasticity. Front Cell Dev Biol 2022; 10:901026. [PMID: 35859899 PMCID: PMC9289519 DOI: 10.3389/fcell.2022.901026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/10/2022] [Indexed: 01/01/2023] Open
Abstract
Collective cell migration on extracellular matrix (ECM) networks is a key biological process involved in development, tissue homeostasis and diseases such as metastatic cancer. During invasion of epithelial cancers, cell clusters migrate through the surrounding stroma, which is comprised primarily of networks of collagen-I fibers. There is growing evidence that the rheological and topological properties of collagen networks can impact cell behavior and cell migration dynamics. During migration, cells exert mechanical forces on their substrate, resulting in an active remodeling of ECM networks that depends not only on the forces produced, but also on the molecular mechanisms that dictate network rheology. One aspect of collagen network rheology whose role is emerging as a crucial parameter in dictating cell behavior is network viscoelasticity. Dynamic reorganization of ECM networks can induce local changes in network organization and mechanics, which can further feed back on cell migration dynamics and cell-cell rearrangement. A number of studies, including many recent publications, have investigated the mechanisms underlying structural changes to collagen networks in response to mechanical force as well as the role of collagen rheology and topology in regulating cell behavior. In this mini-review, we explore the cause-consequence relationship between collagen network viscoelasticity and cell rearrangements at various spatiotemporal scales. We focus on structural alterations of collagen-I networks during collective cell migration and discuss the main rheological parameters, and in particular the role of viscoelasticity, which can contribute to local matrix stiffening during cell movement and can elicit changes in cell dynamics.
Collapse
Affiliation(s)
| | - Milan Milivojevic
- University of Belgrade, Faculty of Technology and Metallurgy, Belgrade, Serbia
| | - Andrew G. Clark
- University of Stuttgart, Institute of Cell Biology and Immunology, Stuttgart, Germany
- University of Stuttgart, Stuttgart Research Center Systems Biology, Stuttgart, Germany
- University of Tübingen, Center for Personalized Medicine, Tübingen, Germany
| |
Collapse
|
100
|
Han W, He M, Zhang Y, Zhou J, Li Z, Liu X, Sun X, Yin X, Yao D, Liang H. Cadherin-dependent adhesion modulated 3D cell-assembly. J Mater Chem B 2022; 10:4959-4966. [PMID: 35730726 DOI: 10.1039/d2tb01006b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The emergence of synthetic biology has opened new avenues in constructing cell-assembly biosystems with specific gene expression and function. The phenomena of cell spreading and detachment during tissue development and cancer metastasis are caused by surface tension, which in turn results from differences in cell-cell adhesion mediated by the dimerization of cadherin expressed on the cell surface. In this study, E- and P-cadherin plasmids were first constructed based on the differential adhesion hypothesis, then they were electroporated into K562 cells and HEK293T cells, respectively, to explore the process of cell migration and assembly regulated by cadherins. Using this approach, some special 3D cell functional components with a phase separation structure were fabricated successfully. Our work will be of potential application in the construction of self-assembling synthetic tissues and organoids.
Collapse
Affiliation(s)
- Wenjie Han
- Hefei National Research Center for Physical Sciences at the Microscale, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Miao He
- School of Chemistry and Materials Science, Department of Polymer Science and Engineering, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Yunhan Zhang
- School of Chemistry and Materials Science, Department of Polymer Science and Engineering, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Junxiang Zhou
- School of Chemistry and Materials Science, Department of Polymer Science and Engineering, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Zhigang Li
- School of Chemistry and Materials Science, Department of Polymer Science and Engineering, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Xiaoyu Liu
- School of Chemistry and Materials Science, Department of Polymer Science and Engineering, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Xiaoyun Sun
- Hefei National Research Center for Physical Sciences at the Microscale, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Xue Yin
- School of Chemistry and Materials Science, Department of Polymer Science and Engineering, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Dongbao Yao
- School of Chemistry and Materials Science, Department of Polymer Science and Engineering, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| | - Haojun Liang
- Hefei National Research Center for Physical Sciences at the Microscale, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China. .,School of Chemistry and Materials Science, Department of Polymer Science and Engineering, iChEM (Collaborative Innovation Center of Chemistry for Energy Materials), University of Science and Technology of China, Hefei, Anhui 230026, China.
| |
Collapse
|