1
|
Vela-Alcántara AM, Santiago-García J, Barragán-Palacios M, León-Chacón A, Domínguez-Pantoja M, Barceinas-Dávila I, Juárez-Aguilar E, Tamariz E. Differential modulation of cell morphology, migration, and Neuropilin-1 expression in cancer and non-cancer cell lines by substrate stiffness. Front Cell Dev Biol 2024; 12:1352233. [PMID: 38903533 PMCID: PMC11188430 DOI: 10.3389/fcell.2024.1352233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/10/2024] [Indexed: 06/22/2024] Open
Abstract
Physical changes in the tumor microenvironment, such as increased stiffness, regulate cancer hallmarks and play an essential role in gene expression, cell morphology, migration, and malignancy. However, the response of cancer cells to stiffness is not homogeneous and varies depending on the cell type and its mechanosensitivity. In this study, we investigated the differential responses of cervical (HeLa) and prostate (PC-3) cancer cell lines, as well as non-tumoral cell lines (HEK293 and HPrEC), to stiffness using polyacrylamide hydrogels mimicking normal and tumoral tissues. We analyzed cell morphology, migration, and the expression of neuropilin 1 (NRP1), a receptor involved in angiogenesis, cell migration, and extracellular matrix remodeling, known to be associated with cancer progression and poor prognosis. Our findings reveal that NRP1 expression increases on substrates mimicking the high stiffness characteristic of tumoral tissue in the non-tumoral cell lines HPrEC and HEK293. Conversely, in tumoral PC-3 cells, stiffness resembling normal prostate tissue induces an earlier and more sustained expression of NRP1. Furthermore, we observed that stiffness influences cell spreading, pseudopodia formation, and the mode of cell protrusion during migration. Soft substrates predominantly trigger bleb cell protrusion, while pseudopodia protrusions increase on substrates mimicking normal and tumor-like stiffnesses in HPrEC cells compared to PC-3 cells. Stiffer substrates also enhance the percentage of migratory cells, as well as their velocity and total displacement, in both non-tumoral and tumoral prostate cells. However, they only improve the persistence of migration in tumoral PC-3 cells. Moreover, we found that NRP1 co-localizes with actin, and its suppression impairs tumoral PC-3 spreading while decreasing pseudopodia protrusion mode. Our results suggest that the modulation of NRP1 expression by the stiffness can be a feedback loop to promote malignancy in non-tumoral and cancer cells, contingent upon the mechanosensitivity of the cells.
Collapse
Affiliation(s)
- Ana Monserrat Vela-Alcántara
- Programa de Doctorado en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Juan Santiago-García
- Laboratorio de Biología Molecular, Instituto de Investigaciones Biológicas, Universidad Veracruzana, Xalapa, Mexico
| | - Madeleine Barragán-Palacios
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Programa de Maestría en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Aylin León-Chacón
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | | | - Irene Barceinas-Dávila
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
- Programa de Maestría en Ciencias de la Salud, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Enrique Juárez-Aguilar
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| | - Elisa Tamariz
- Laboratorio de Cultivo Celular, Departamento de Biomedicina, Instituto de Ciencias de la Salud, Universidad Veracruzana, Xalapa, Mexico
| |
Collapse
|
2
|
Mierke CT. Phenotypic Heterogeneity, Bidirectionality, Universal Cues, Plasticity, Mechanics, and the Tumor Microenvironment Drive Cancer Metastasis. Biomolecules 2024; 14:184. [PMID: 38397421 PMCID: PMC10887446 DOI: 10.3390/biom14020184] [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: 12/25/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor diseases become a huge problem when they embark on a path that advances to malignancy, such as the process of metastasis. Cancer metastasis has been thoroughly investigated from a biological perspective in the past, whereas it has still been less explored from a physical perspective. Until now, the intraluminal pathway of cancer metastasis has received the most attention, while the interaction of cancer cells with macrophages has received little attention. Apart from the biochemical characteristics, tumor treatments also rely on the tumor microenvironment, which is recognized to be immunosuppressive and, as has recently been found, mechanically stimulates cancer cells and thus alters their functions. The review article highlights the interaction of cancer cells with other cells in the vascular metastatic route and discusses the impact of this intercellular interplay on the mechanical characteristics and subsequently on the functionality of cancer cells. For instance, macrophages can guide cancer cells on their intravascular route of cancer metastasis, whereby they can help to circumvent the adverse conditions within blood or lymphatic vessels. Macrophages induce microchannel tunneling that can possibly avoid mechanical forces during extra- and intravasation and reduce the forces within the vascular lumen due to vascular flow. The review article highlights the vascular route of cancer metastasis and discusses the key players in this traditional route. Moreover, the effects of flows during the process of metastasis are presented, and the effects of the microenvironment, such as mechanical influences, are characterized. Finally, the increased knowledge of cancer metastasis opens up new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
3
|
Mierke CT. Magnetic tweezers in cell mechanics. Methods Enzymol 2024; 694:321-354. [PMID: 38492957 DOI: 10.1016/bs.mie.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2024]
Abstract
The chapter provides an overview of the applications of magnetic tweezers in living cells. It discusses the advantages and disadvantages of magnetic tweezers technology with a focus on individual magnetic tweezers configurations, such as electromagnetic tweezers. Solutions to the disadvantages identified are also outlined. The specific role of magnetic tweezers in the field of mechanobiology, such as mechanosensitivity, mechano-allostery and mechanotransduction are also emphasized. The specific usage of magnetic tweezers in mechanically probing cells via specific cell surface receptors, such as mechanosensitive channels is discussed and why mechanical probing has revealed the opening and closing of the channels. Finally, the future direction of magnetic tweezers is presented.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Sciences, Peter Debye Institute for Soft Matter Physics, Biological Physics Division, Leipzig University, Leipzig, Germany.
| |
Collapse
|
4
|
Bozzuto G, Colone M, Toccacieli L, Molinari A, Calcabrini A, Stringaro A. Interaction of Drug-Sensitive and -Resistant Human Melanoma Cells with HUVEC Cells: A Label-Free Cell-Based Impedance Study. Biomedicines 2023; 11:1544. [PMID: 37371639 DOI: 10.3390/biomedicines11061544] [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: 04/24/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Cancer cell extravasation is a crucial step in cancer metastasis. However, many of the mechanisms involved in this process are only now being elucidated. Thus, in the present study we analysed the trans-endothelial invasion of melanoma cells by a high throughput label-free cell impedance assay applied to transwell chamber invasion assay. This technique monitors and quantifies in real-time the invasion of endothelial cells by malignant tumour cells, for a long time, avoiding artefacts due to preparation of the end point measurements. Results obtained by impedance analysis were compared with endpoint measurements. In this study, we used human melanoma M14 wild type (WT) cells and their drug resistant counterparts, M14 multidrug resistant (ADR) melanoma cells, selected by prolonged exposure to doxorubicin (DOX). Tumour cells were co-cultured with monolayers of human umbilical vein endothelial cells (HUVEC). Results herein reported demonstrated that: (i) the trans-endothelial migration of resistant melanoma cells was faster than sensitive ones; (ii) the endothelial cells appeared to be strongly affected by the transmigration of melanoma cells which showed the ability to degrade their cytoplasm; (iii) resistant cells preferentially adopted the transcellular invasion vs. the paracellular one; (iv) the endothelial damage mediated by tumour metalloproteinases seemed to be reversible.
Collapse
Affiliation(s)
- Giuseppina Bozzuto
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marisa Colone
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Laura Toccacieli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Agnese Molinari
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Annarica Calcabrini
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Annarita Stringaro
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
5
|
Xie L, Sun Z, Brown NJ, Glinskii OV, Meininger GA, Glinsky VV. Changes in dynamics of tumor/endothelial cell adhesive interactions depending on endothelial cell growth state and elastic properties. PLoS One 2022; 17:e0269552. [PMID: 35666755 PMCID: PMC9170101 DOI: 10.1371/journal.pone.0269552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer cell adhesion to the endothelium is a crucial process in hematogenous metastasis, but how the integrity of the endothelial barrier and endothelial cell (EC) mechanical properties influence the adhesion between metastatic cancer cells and the endothelium remain unclear. In the present study, we have measured the adhesion between single cancer cells and two types of ECs at various growth states and their mechanical properties (elasticity) using atomic force microscopy single cell force spectroscopy. We demonstrated that the EC stiffness increased and adhesion with cancer cells decreased, as ECs grew from a single cell to a confluent state and developed cell-cell contacts, but this was reversed when confluent cells returned to a single state in a scratch assay. Our results suggest that the integrity of the endothelial barrier is an important factor in reducing the ability of the metastatic tumor cells to adhere to the vascular endothelium, extravasate and lodge in the vasculature of a distant organ where secondary metastatic tumors would develop.
Collapse
Affiliation(s)
- Leike Xie
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
| | - Zhe Sun
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
| | - Nicola J. Brown
- Microcirculation Research Group, Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, United Kingdom
| | - Olga V. Glinskii
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States of America
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States of America
| | - Gerald A. Meininger
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, United States of America
| | - Vladislav V. Glinsky
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, United States of America
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, United States of America
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States of America
| |
Collapse
|
6
|
Koszalka P, Kutryb-Zajac B, Mierzejewska P, Tomczyk M, Wietrzyk J, Serafin PK, Smolenski RT, Slominska EM. 4-Pyridone-3-carboxamide-1-β-D-ribonucleoside (4PYR)—A Novel Oncometabolite Modulating Cancer-Endothelial Interactions in Breast Cancer Metastasis. Int J Mol Sci 2022; 23:ijms23105774. [PMID: 35628582 PMCID: PMC9145394 DOI: 10.3390/ijms23105774] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
The accumulation of specific metabolic intermediates is known to promote cancer progression. We analyzed the role of 4-pyridone-3-carboxamide-1-β-D-ribonucleoside (4PYR), a nucleotide metabolite that accumulates in the blood of cancer patients, using the 4T1 murine in vivo breast cancer model, and cultured cancer (4T1) and endothelial cells (ECs) for in vitro studies. In vivo studies demonstrated that 4PYR facilitated lung metastasis without affecting primary tumor growth. In vitro studies demonstrated that 4PYR affected extracellular adenine nucleotide metabolism and the intracellular energy status in ECs, shifting catabolite patterns toward the accumulation of extracellular inosine, and leading to the increased permeability of lung ECs. These changes prevailed over the direct effect of 4PYR on 4T1 cells that reduced their invasive potential through 4PYR-induced modulation of the CD73-adenosine axis. We conclude that 4PYR is an oncometabolite that affects later stages of the metastatic cascade by acting specifically through the regulation of EC permeability and metabolic controls of inflammation.
Collapse
Affiliation(s)
- Patrycja Koszalka
- Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdansk, 80-210 Gdansk, Poland;
- Correspondence: (P.K.); (E.M.S.); Tel.: +48-58-349-1410 (P.K.); +48-58-349-1006 (E.M.S.)
| | - Barbara Kutryb-Zajac
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Paulina Mierzejewska
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Marta Tomczyk
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wroclaw, Poland;
| | - Pawel K. Serafin
- Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdansk, 80-210 Gdansk, Poland;
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
| | - Ewa M. Slominska
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (B.K.-Z.); (P.M.); (M.T.); (R.T.S.)
- Correspondence: (P.K.); (E.M.S.); Tel.: +48-58-349-1410 (P.K.); +48-58-349-1006 (E.M.S.)
| |
Collapse
|
7
|
Mierke CT. Viscoelasticity, Like Forces, Plays a Role in Mechanotransduction. Front Cell Dev Biol 2022; 10:789841. [PMID: 35223831 PMCID: PMC8864183 DOI: 10.3389/fcell.2022.789841] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Viscoelasticity and its alteration in time and space has turned out to act as a key element in fundamental biological processes in living systems, such as morphogenesis and motility. Based on experimental and theoretical findings it can be proposed that viscoelasticity of cells, spheroids and tissues seems to be a collective characteristic that demands macromolecular, intracellular component and intercellular interactions. A major challenge is to couple the alterations in the macroscopic structural or material characteristics of cells, spheroids and tissues, such as cell and tissue phase transitions, to the microscopic interferences of their elements. Therefore, the biophysical technologies need to be improved, advanced and connected to classical biological assays. In this review, the viscoelastic nature of cytoskeletal, extracellular and cellular networks is presented and discussed. Viscoelasticity is conceptualized as a major contributor to cell migration and invasion and it is discussed whether it can serve as a biomarker for the cells' migratory capacity in several biological contexts. It can be hypothesized that the statistical mechanics of intra- and extracellular networks may be applied in the future as a powerful tool to explore quantitatively the biomechanical foundation of viscoelasticity over a broad range of time and length scales. Finally, the importance of the cellular viscoelasticity is illustrated in identifying and characterizing multiple disorders, such as cancer, tissue injuries, acute or chronic inflammations or fibrotic diseases.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
8
|
Herbig M, Jacobi A, Wobus M, Weidner H, Mies A, Kräter M, Otto O, Thiede C, Weickert MT, Götze KS, Rauner M, Hofbauer LC, Bornhäuser M, Guck J, Ader M, Platzbecker U, Balaian E. Machine learning assisted real-time deformability cytometry of CD34+ cells allows to identify patients with myelodysplastic syndromes. Sci Rep 2022; 12:870. [PMID: 35042906 PMCID: PMC8766444 DOI: 10.1038/s41598-022-04939-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 01/03/2022] [Indexed: 12/13/2022] Open
Abstract
Diagnosis of myelodysplastic syndrome (MDS) mainly relies on a manual assessment of the peripheral blood and bone marrow cell morphology. The WHO guidelines suggest a visual screening of 200 to 500 cells which inevitably turns the assessor blind to rare cell populations and leads to low reproducibility. Moreover, the human eye is not suited to detect shifts of cellular properties of entire populations. Hence, quantitative image analysis could improve the accuracy and reproducibility of MDS diagnosis. We used real-time deformability cytometry (RT-DC) to measure bone marrow biopsy samples of MDS patients and age-matched healthy individuals. RT-DC is a high-throughput (1000 cells/s) imaging flow cytometer capable of recording morphological and mechanical properties of single cells. Properties of single cells were quantified using automated image analysis, and machine learning was employed to discover morpho-mechanical patterns in thousands of individual cells that allow to distinguish healthy vs. MDS samples. We found that distribution properties of cell sizes differ between healthy and MDS, with MDS showing a narrower distribution of cell sizes. Furthermore, we found a strong correlation between the mechanical properties of cells and the number of disease-determining mutations, inaccessible with current diagnostic approaches. Hence, machine-learning assisted RT-DC could be a promising tool to automate sample analysis to assist experts during diagnosis or provide a scalable solution for MDS diagnosis to regions lacking sufficient medical experts.
Collapse
Affiliation(s)
- Maik Herbig
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Angela Jacobi
- Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
- Medical Department I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Manja Wobus
- Medical Department I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Heike Weidner
- Medical Department III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
- Center for Healthy Aging, Dresden, Germany
| | - Anna Mies
- Medical Department I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Oliver Otto
- Zentrum für Innovationskompetenz: Humorale Immunreaktionen in Kardiovaskulären Erkrankungen, Universität Greifswald, Greifswald, Germany
| | - Christian Thiede
- Medical Department I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
| | - Marie-Theresa Weickert
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Katharina S Götze
- Department of Medicine III: Hematology and Oncology, School of Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Martina Rauner
- Medical Department III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
- Center for Healthy Aging, Dresden, Germany
| | - Lorenz C Hofbauer
- Medical Department III, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Center for Healthy Aging, Dresden, Germany
| | - Martin Bornhäuser
- Medical Department I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Marius Ader
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Uwe Platzbecker
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Cellular Therapy and Hemostaseology, Leipzig University Hospital, Leipzig, Germany
| | - Ekaterina Balaian
- Medical Department I, University Hospital Carl Gustav Carus Dresden, Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
9
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 294] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
10
|
Fei L, Ren X, Yu H, Zhan Y. Targeting the CCL2/CCR2 Axis in Cancer Immunotherapy: One Stone, Three Birds? Front Immunol 2021; 12:771210. [PMID: 34804061 PMCID: PMC8596464 DOI: 10.3389/fimmu.2021.771210] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/19/2021] [Indexed: 12/15/2022] Open
Abstract
CCR2 is predominantly expressed by monocytes/macrophages with strong proinflammatory functions, prompting the development of CCR2 antagonists to dampen unwanted immune responses in inflammatory and autoimmune diseases. Paradoxically, CCR2-expressing monocytes/macrophages, particularly in tumor microenvironments, can be strongly immunosuppressive. Thus, targeting the recruitment of immunosuppressive monocytes/macrophages to tumors by CCR2 antagonism has recently been investigated as a strategy to modify the tumor microenvironment and enhance anti-tumor immunity. We present here that beneficial effects of CCR2 antagonism in the tumor setting extend beyond blocking chemotaxis of suppressive myeloid cells. Signaling within the CCL2/CCR2 axis shows underappreciated effects on myeloid cell survival and function polarization. Apart from myeloid cells, T cells are also known to express CCR2. Nevertheless, tissue homing of Treg cells among T cell populations is preferentially affected by CCR2 deficiency. Further, CCR2 signaling also directly enhances Treg functional potency. Thus, although Tregs are not the sole type of T cells expressing CCR2, the net outcome of CCR2 antagonism in T cells favors the anti-tumor arm of immune responses. Finally, the CCL2/CCR2 axis directly contributes to survival/growth and invasion/metastasis of many types of tumors bearing CCR2. Together, CCR2 links to two main types of suppressive immune cells by multiple mechanisms. Such a CCR2-assoicated immunosuppressive network is further entangled with paracrine and autocrine CCR2 signaling of tumor cells. Strategies to target CCL2/CCR2 axis as cancer therapy in the view of three types of CCR2-expessing cells in tumor microenvironment are discussed.
Collapse
Affiliation(s)
- Liyang Fei
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Xiaochen Ren
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Haijia Yu
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Yifan Zhan
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| |
Collapse
|
11
|
Mierke CT. The Pertinent Role of Cell and Matrix Mechanics in Cell Adhesion and Migration. Front Cell Dev Biol 2021; 9:720494. [PMID: 34722504 PMCID: PMC8548417 DOI: 10.3389/fcell.2021.720494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/20/2021] [Indexed: 01/17/2023] Open
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
12
|
Control of Tumor Progression by Angiocrine Factors. Cancers (Basel) 2021; 13:cancers13112610. [PMID: 34073394 PMCID: PMC8198241 DOI: 10.3390/cancers13112610] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/24/2022] Open
Abstract
Tumor progression, therapy resistance and metastasis are profoundly controlled by the tumor microenvironment. The contribution of endothelial cells to tumor progression was initially only attributed to the formation of new blood vessels (angiogenesis). Research in the last decade has revealed however that endothelial cells control their microenvironment through the expression of membrane-bound and secreted factors. Such angiocrine functions are frequently hijacked by cancer cells, which deregulate the signaling pathways controlling the expression of angiocrine factors. Here, we review the crosstalk between cancer cells and endothelial cells and how this contributes to the cancer stem cell phenotype, epithelial to mesenchymal transition, immunosuppression, remodeling of the extracellular matrix and intravasation of cancer cells into the bloodstream. We also address the long-distance crosstalk of a primary tumor with endothelial cells at the pre-metastatic niche and how this contributes to metastasis.
Collapse
|
13
|
Aermes C, Hayn A, Fischer T, Mierke CT. Cell mechanical properties of human breast carcinoma cells depend on temperature. Sci Rep 2021; 11:10771. [PMID: 34031462 PMCID: PMC8144563 DOI: 10.1038/s41598-021-90173-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022] Open
Abstract
The knowledge of cell mechanics is required to understand cellular processes and functions, such as the movement of cells, and the development of tissue engineering in cancer therapy. Cell mechanical properties depend on a variety of factors, such as cellular environments, and may also rely on external factors, such as the ambient temperature. The impact of temperature on cell mechanics is not clearly understood. To explore the effect of temperature on cell mechanics, we employed magnetic tweezers to apply a force of 1 nN to 4.5 µm superparamagnetic beads. The beads were coated with fibronectin and coupled to human epithelial breast cancer cells, in particular MCF-7 and MDA-MB-231 cells. Cells were measured in a temperature range between 25 and 45 °C. The creep response of both cell types followed a weak power law. At all temperatures, the MDA-MB-231 cells were pronouncedly softer compared to the MCF-7 cells, whereas their fluidity was increased. However, with increasing temperature, the cells became significantly softer and more fluid. Since mechanical properties are manifested in the cell's cytoskeletal structure and the paramagnetic beads are coupled through cell surface receptors linked to cytoskeletal structures, such as actin and myosin filaments as well as microtubules, the cells were probed with pharmacological drugs impacting the actin filament polymerization, such as Latrunculin A, the myosin filaments, such as Blebbistatin, and the microtubules, such as Demecolcine, during the magnetic tweezer measurements in the specific temperature range. Irrespective of pharmacological interventions, the creep response of cells followed a weak power law at all temperatures. Inhibition of the actin polymerization resulted in increased softness in both cell types and decreased fluidity exclusively in MDA-MB-231 cells. Blebbistatin had an effect on the compliance of MDA-MB-231 cells at lower temperatures, which was minor on the compliance MCF-7 cells. Microtubule inhibition affected the fluidity of MCF-7 cells but did not have a significant effect on the compliance of MCF-7 and MDA-MB-231 cells. In summary, with increasing temperature, the cells became significant softer with specific differences between the investigated drugs and cell lines.
Collapse
Affiliation(s)
- Christian Aermes
- Biological Physics Division, Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, University of Leipzig, Linnéstr. 5, 04103, Leipzig, Germany
| | - Alexander Hayn
- Biological Physics Division, Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, University of Leipzig, Linnéstr. 5, 04103, Leipzig, Germany
| | - Tony Fischer
- Biological Physics Division, Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, University of Leipzig, Linnéstr. 5, 04103, Leipzig, Germany
| | - Claudia Tanja Mierke
- Biological Physics Division, Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, University of Leipzig, Linnéstr. 5, 04103, Leipzig, Germany.
| |
Collapse
|
14
|
Nanou A, Bourbouli M, Vetrano S, Schaeper U, Ley S, Kollias G. Endothelial Tpl2 regulates vascular barrier function via JNK-mediated degradation of claudin-5 promoting neuroinflammation or tumor metastasis. Cell Rep 2021; 35:109168. [PMID: 34038728 DOI: 10.1016/j.celrep.2021.109168] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 04/08/2021] [Accepted: 05/03/2021] [Indexed: 12/15/2022] Open
Abstract
Increased vascular permeability and leakage are hallmarks of several pathologies and determine disease progression and severity by facilitating inflammatory/metastatic cell infiltration. Using tissue-specific genetic ablation in endothelial cells, we have investigated in vivo the role of Tumor progression locus 2 (Tpl2), a mitogen-activated protein kinase kinase kinase (MAP3K) member with pleiotropic effects in inflammation and cancer. In response to proinflammatory stimuli, endothelial Tpl2 deletion alters tight junction claudin-5 protein expression through inhibition of JNK signaling and lysosomal degradation activation, resulting in reduced vascular permeability and immune cell infiltration. This results in significantly attenuated disease scores in experimental autoimmune encephalomyelitis and fewer tumor nodules in a hematogenic lung cancer metastasis model. Accordingly, pharmacologic inhibition of Tpl2 or small interfering RNA (siRNA)-mediated Tpl2 knockdown recapitulates our findings and reduces lung metastatic tumor invasions. These results establish an endothelial-specific role for Tpl2 and highlight the therapeutic potential of blocking the endothelial-specific Tpl2 pathway in chronic inflammatory and metastatic diseases.
Collapse
Affiliation(s)
- Aikaterini Nanou
- Institute for Bioinnovation, Biomedical Science Research Center (BSRC) "Alexander Fleming," Vari, Attika, Greece
| | - Mara Bourbouli
- Institute for Bioinnovation, Biomedical Science Research Center (BSRC) "Alexander Fleming," Vari, Attika, Greece
| | - Stefania Vetrano
- Department of Biomedical Sciences, Humanitas University, Rozzano, Italy; IBD Center, Humanitas Research Hospital, Rozzano, Italy
| | | | - Steven Ley
- Immune Cell Signalling Laboratory, The Francis Crick Institute, London, UK; Imperial College, London, UK
| | - George Kollias
- Institute for Bioinnovation, Biomedical Science Research Center (BSRC) "Alexander Fleming," Vari, Attika, Greece; Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
15
|
Nie Y, Jiang MC, Liu C, Liu Q, Zhu X. CXCL5 Has Potential to Be a Marker for Hepatocellular Carcinoma Prognosis and Was Correlating With Immune Infiltrates. Front Oncol 2021; 11:637023. [PMID: 33869023 PMCID: PMC8045759 DOI: 10.3389/fonc.2021.637023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/01/2021] [Indexed: 01/10/2023] Open
Abstract
Backgrounds Tumor microenvironment (TME) plays a crucial role in the initiation and progression of Hepatocellular Carcinoma (HCC), especially immune infiltrates. However, there is still a challenge in understanding the modulation of the immune and stromal components in TME, especially TME related genes. Methods The proportion of tumor-infiltrating immune cells (TICs) and the immune and stromal scores in 374 HCC patients from The Cancer Genome Atlas (TCGA) database were determined using CIBERSORT and ESTIMATE computational methods. The final screened genes were confirmed by the PPI network and univariate Cox regression of the differentially expressed genes based on different immune or stromal scores. The correlation between the expression levels of the final gene interactions and the clinical characteristics was based on TCGA database and local hospital data. Gene set enrichment analysis (GSEA) and the effect of CXCL5 expression on TICs were conducted. Results There were correlations between the expression of CXCL5 and survival of HCC patients and TMN classification both in TCGA database and local hospital data. The immune-related activities were enriched in the high-expression group; however, the metabolic pathways were enriched in the low-expression group. The result of CIBERSORT analyzing had indicated that CXCL5 expression were correlated with the proportion of NK cells activated, macrophages M0, Mast cells resting, Neutrophils. Conclusions CXCL5 was a potential prognostic marker for HCC and provides clues regarding immune infiltrates, which offers extra insight for therapeutics of HCC, however, more independent cohorts and functional experiments of CXCL5 are warranted.
Collapse
Affiliation(s)
- Yuan Nie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mei-Chun Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Cong Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qi Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
16
|
Hilfenhaus G, Mompeón A, Freshman J, Prajapati DP, Hernandez G, Freitas VM, Ma F, Langenbacher AD, Mirkov S, Song D, Cho BK, Goo YA, Pellegrini M, Chen JN, Damoiseaux R, Iruela-Arispe ML. A High-Content Screen Identifies Drugs That Restrict Tumor Cell Extravasation across the Endothelial Barrier. Cancer Res 2020; 81:619-633. [PMID: 33218969 DOI: 10.1158/0008-5472.can-19-3911] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 09/11/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022]
Abstract
Metastases largely rely on hematogenous dissemination of tumor cells via the vascular system and significantly limit prognosis of patients with solid tumors. To colonize distant sites, circulating tumor cells must destabilize the endothelial barrier and transmigrate across the vessel wall. Here we performed a high-content screen to identify drugs that block tumor cell extravasation by testing 3,520 compounds on a transendothelial invasion coculture assay. Hits were further characterized and validated using a series of in vitro assays, a zebrafish model enabling three-dimensional (3D) visualization of tumor cell extravasation, and mouse models of lung metastasis. The initial screen advanced 38 compounds as potential hits, of which, four compounds enhanced endothelial barrier stability while concurrently suppressing tumor cell motility. Two compounds niclosamide and forskolin significantly reduced tumor cell extravasation in zebrafish, and niclosamide drastically impaired metastasis in mice. Because niclosamide had not previously been linked with effects on barrier function, single-cell RNA sequencing uncovered mechanistic effects of the drug on both tumor and endothelial cells. Importantly, niclosamide affected homotypic and heterotypic signaling critical to intercellular junctions, cell-matrix interactions, and cytoskeletal regulation. Proteomic analysis indicated that niclosamide-treated mice also showed reduced levels of kininogen, the precursor to the permeability mediator bradykinin. Our findings designate niclosamide as an effective drug that restricts tumor cell extravasation through modulation of signaling pathways, chemokines, and tumor-endothelial cell interactions. SIGNIFICANCE: A high-content screen identified niclosamide as an effective drug that restricts tumor cell extravasation by enhancing endothelial barrier stability through modulation of molecular signaling, chemokines, and tumor-endothelial cell interactions. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/3/619/F1.large.jpg.
Collapse
Affiliation(s)
- Georg Hilfenhaus
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Ana Mompeón
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jonathan Freshman
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Divya P Prajapati
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Gloria Hernandez
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Vanessa M Freitas
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Feiyang Ma
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Adam D Langenbacher
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Snezana Mirkov
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Dana Song
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
| | - Byoung-Kyu Cho
- Proteomics Center of Excellence, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Young Ah Goo
- Proteomics Center of Excellence, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Jau-Nian Chen
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - M Luisa Iruela-Arispe
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California.
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
17
|
Mierke CT. Mechanical Cues Affect Migration and Invasion of Cells From Three Different Directions. Front Cell Dev Biol 2020; 8:583226. [PMID: 33043017 PMCID: PMC7527720 DOI: 10.3389/fcell.2020.583226] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Cell migration and invasion is a key driving factor for providing essential cellular functions under physiological conditions or the malignant progression of tumors following downward the metastatic cascade. Although there has been plentiful of molecules identified to support the migration and invasion of cells, the mechanical aspects have not yet been explored in a combined and systematic manner. In addition, the cellular environment has been classically and frequently assumed to be homogeneous for reasons of simplicity. However, motility assays have led to various models for migration covering only some aspects and supporting factors that in some cases also include mechanical factors. Instead of specific models, in this review, a more or less holistic model for cell motility in 3D is envisioned covering all these different aspects with a special emphasis on the mechanical cues from a biophysical perspective. After introducing the mechanical aspects of cell migration and invasion and presenting the heterogeneity of extracellular matrices, the three distinct directions of cell motility focusing on the mechanical aspects are presented. These three different directions are as follows: firstly, the commonly used invasion tests using structural and structure-based mechanical environmental signals; secondly, the mechano-invasion assay, in which cells are studied by mechanical forces to migrate and invade; and thirdly, cell mechanics, including cytoskeletal and nuclear mechanics, to influence cell migration and invasion. Since the interaction between the cell and the microenvironment is bi-directional in these assays, these should be accounted in migration and invasion approaches focusing on the mechanical aspects. Beyond this, there is also the interaction between the cytoskeleton of the cell and its other compartments, such as the cell nucleus. In specific, a three-element approach is presented for addressing the effect of mechanics on cell migration and invasion by including the effect of the mechano-phenotype of the cytoskeleton, nucleus and the cell's microenvironment into the analysis. In precise terms, the combination of these three research approaches including experimental techniques seems to be promising for revealing bi-directional impacts of mechanical alterations of the cellular microenvironment on cells and internal mechanical fluctuations or changes of cells on the surroundings. Finally, different approaches are discussed and thereby a model for the broad impact of mechanics on cell migration and invasion is evolved.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
18
|
Aermes C, Hayn A, Fischer T, Mierke CT. Environmentally controlled magnetic nano-tweezer for living cells and extracellular matrices. Sci Rep 2020; 10:13453. [PMID: 32778758 PMCID: PMC7417586 DOI: 10.1038/s41598-020-70428-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 07/16/2020] [Indexed: 01/08/2023] Open
Abstract
The magnetic tweezer technique has become a versatile tool for unfolding or folding of individual molecules, mainly DNA. In addition to single molecule analysis, the magnetic tweezer can be used to analyze the mechanical properties of cells and extracellular matrices. We have established a magnetic tweezer that is capable of measuring the linear and non-linear viscoelastic behavior of a wide range of soft matter in precisely controlled environmental conditions, such as temperature, CO2 and humidity. The magnetic tweezer presented in this study is suitable to detect specific differences in the mechanical properties of different cell lines, such as human breast cancer cells and mouse embryonic fibroblasts, as well as collagen matrices of distinct concentrations in the presence and absence of fibronectin crosslinks. The precise calibration and control mechanism employed in the presented magnetic tweezer setup provides the ability to apply physiological force up to 5 nN on 4.5 µm superparamagnetic beads coated with fibronectin and coupled to the cells or collagen matrices. These measurements reveal specific local linear and non-linear viscoelastic behavior of the investigated samples. The viscoelastic response of cells and collagen matrices to the force application is best described by a weak power law behavior. Our results demonstrate that the stress stiffening response and the fluidization of cells is cell type specific and varies largely between differently invasive and aggressive cancer cells. Finally, we showed that the viscoelastic behavior of collagen matrices with and without fibronectin crosslinks measured by the magnetic tweezer can be related to the microstructure of these matrices.
Collapse
Affiliation(s)
- Christian Aermes
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Linnéstr. 5, 04103, Leipzig, Germany
| | - Alexander Hayn
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Linnéstr. 5, 04103, Leipzig, Germany
| | - Tony Fischer
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Linnéstr. 5, 04103, Leipzig, Germany
| | - Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Linnéstr. 5, 04103, Leipzig, Germany.
| |
Collapse
|
19
|
Zamani ARN, Avci ÇB, Ahmadi M, Pouyafar A, Bagheri HS, Fathi F, Heidarzadeh M, Rezaie J, Mirhosseini Y, Saberianpour S, Mehdizadeh A, Sokullu E, Talebi M, Rahbarghazi R. Estradiol modulated colorectal cancer stem cells bioactivity and interaction with endothelial cells. Life Sci 2020; 257:118078. [PMID: 32663577 DOI: 10.1016/j.lfs.2020.118078] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022]
Abstract
This study aimed to evaluate the modulatory role of sex-related hormone estradiol on cancer stem cells with the origin of colorectal adenocarcinoma in vitro. Cancer stem cells were incubated with 100 nM estradiol for 48 h. The cell survival rate was analyzed using the MTT assay. Immunocytochemistry staining of Ki-67 and Inhibin and Apoptosis PCR array were done to measure proliferation/apoptosis. Cell migration was monitored via the Transwell Migration assay. The expression of exosome biogenesis genes was measured using a real-time PCR assay. The fatty acid profile was monitored using gas chromatography. The level of FAK, SQSTM1, ER, and SIRT1 was examined using Western blotting. Cancer stem-endothelial cell interaction was investigated using Surface Plasmon Resonance assay. Data showed no significant differences in cancer stem cell viability and proliferation between control and estradiol-treated groups (p>0.05). PCR array highlighted the up-regulation of both pro- and anti-apoptosis effectors in the treatment group compared to the control cells (p<0.05). Cell migration capacity was increased after treatment with estradiol (p<0.001). Both exocytosis and exosome biogenesis were decreased in cancer stem cells exposed to estradiol (p<0.05). Data showed the reduction of palmitic acid, and increase of Palmitoleic and Linolenic acids in estradiol-treated cells. Estrogen induced estrogen receptor, SQSTM1 proteins and decreased SIRT1 factor after 48 h. Surface Plasmon Resonance revealed the suppression of cancer stem-endothelial cell interaction and affinity. Estradiol could change the migration, juxtacrine and paracrine activities of cancer stem cells, showing the importance of sex-related hormones in the dynamic of cancer development.
Collapse
Affiliation(s)
| | - Çigir Biray Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Izmir, Turkey
| | - Mahdi Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayda Pouyafar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farzaneh Fathi
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Morteza Heidarzadeh
- Koç University, School of Medicine, Biophysics Department, Rumeli Fener, Sarıyer, Istanbul
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Yasaman Mirhosseini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Saberianpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Emel Sokullu
- Koç University, School of Medicine, Biophysics Department, Rumeli Fener, Sarıyer, Istanbul; Koç University Research Center for Translational Medicine, KUTTAM, Rumeli Feneri Sarıyer, Istanbul, Turkey
| | - Mehdi Talebi
- Hematology And Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Fischer T, Hayn A, Mierke CT. Effect of Nuclear Stiffness on Cell Mechanics and Migration of Human Breast Cancer Cells. Front Cell Dev Biol 2020; 8:393. [PMID: 32548118 PMCID: PMC7272586 DOI: 10.3389/fcell.2020.00393] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 04/29/2020] [Indexed: 12/18/2022] Open
Abstract
The migration and invasion of cancer cells through 3D confined extracellular matrices is coupled to cell mechanics and the mechanics of the extracellular matrix. Cell mechanics is mainly determined by both the mechanics of the largest organelle in the cell, the nucleus, and the cytoskeletal architecture of the cell. Hence, cytoskeletal and nuclear mechanics are the major contributors to cell mechanics. Among other factors, steric hindrances of the extracellular matrix confinement are supposed to affect nuclear mechanics and thus also influence cell mechanics. Therefore, we propose that the percentage of invasive cells and their invasion depths into loose and dense 3D extracellular matrices is regulated by both nuclear and cytoskeletal mechanics. In order to investigate the effect of both nuclear and cytoskeletal mechanics on the overall cell mechanics, we firstly altered nuclear mechanics by the chromatin de-condensing reagent Trichostatin A (TSA) and secondly altered cytoskeletal mechanics by addition of actin polymerization inhibitor Latrunculin A and the myosin inhibitor Blebbistatin. In fact, we found that TSA-treated MDA-MB-231 human breast cancer cells increased their invasion depth in dense 3D extracellular matrices, whereas the invasion depths in loose matrices were decreased. Similarly, the invasion depths of TSA-treated MCF-7 human breast cancer cells in dense matrices were significantly increased compared to loose matrices, where the invasion depths were decreased. These results are also valid in the presence of a matrix-metalloproteinase inhibitor GM6001. Using atomic force microscopy (AFM), we found that the nuclear stiffnesses of both MDA-MB-231 and MCF-7 breast cancer cells were pronouncedly higher than their cytoskeletal stiffness, whereas the stiffness of the nucleus of human mammary epithelial cells was decreased compared to their cytoskeleton. TSA treatment reduced cytoskeletal and nuclear stiffness of MCF-7 cells, as expected. However, a softening of the nucleus by TSA treatment may induce a stiffening of the cytoskeleton of MDA-MB-231 cells and subsequently an apparent stiffening of the nucleus. Inhibiting actin polymerization using Latrunculin A revealed a softer nucleus of MDA-MB-231 cells under TSA treatment. This indicates that the actin-dependent cytoskeletal stiffness seems to be influenced by the TSA-induced nuclear stiffness changes. Finally, the combined treatment with TSA and Latrunculin A further justifies the hypothesis of apparent nuclear stiffening, indicating that cytoskeletal mechanics seem to be regulated by nuclear mechanics.
Collapse
Affiliation(s)
- Tony Fischer
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| | - Alexander Hayn
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| | - Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| |
Collapse
|
21
|
Doolin MT, Moriarty RA, Stroka KM. Mechanosensing of Mechanical Confinement by Mesenchymal-Like Cells. Front Physiol 2020; 11:365. [PMID: 32390868 PMCID: PMC7193100 DOI: 10.3389/fphys.2020.00365] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 03/30/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) and tumor cells have the unique capability to migrate out of their native environment and either home or metastasize, respectively, through extremely heterogeneous environments to a distant location. Once there, they can either aid in tissue regrowth or impart an immunomodulatory effect in the case of MSCs, or form secondary tumors in the case of tumor cells. During these journeys, cells experience physically confining forces that impinge on the cell body and the nucleus, ultimately causing a multitude of cellular changes. Most drastically, confining individual MSCs within hydrogels or confining monolayers of MSCs within agarose wells can sway MSC lineage commitment, while applying a confining compressive stress to metastatic tumor cells can increase their invasiveness. In this review, we seek to understand the signaling cascades that occur as cells sense confining forces and how that translates to behavioral changes, including elongated and multinucleated cell morphologies, novel migrational mechanisms, and altered gene expression, leading to a unique MSC secretome that could hold great promise for anti-inflammatory treatments. Through comparison of these altered behaviors, we aim to discern how MSCs alter their lineage selection, while tumor cells may become more aggressive and invasive. Synthesizing this information can be useful for employing MSCs for therapeutic approaches through systemic injections or tissue engineered grafts, and developing improved strategies for metastatic cancer therapies.
Collapse
Affiliation(s)
- Mary T. Doolin
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
| | - Rebecca A. Moriarty
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD, United States
- Maryland Biophysics Program, University of Maryland, College Park, College Park, MD, United States
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|
22
|
Mun SG, Choi HW, Lee JM, Lim JH, Ha JH, Kang MJ, Kim EJ, Kang L, Chung BG. rGO nanomaterial-mediated cancer targeting and photothermal therapy in a microfluidic co-culture platform. NANO CONVERGENCE 2020; 7:10. [PMID: 32180051 PMCID: PMC7076105 DOI: 10.1186/s40580-020-0220-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/28/2020] [Indexed: 05/15/2023]
Abstract
We developed the microfluidic co-culture platform to study photothermal therapy applications. We conjugated folic acid (FA) to target breast cancer cells using reduced graphene oxide (rGO)-based functional nanomaterials. To characterize the structure of rGO-based nanomaterials, we analyzed the molecular spectrum using UV-visible and Fourier-transform infrared spectroscopy (FT-IR). We demonstrated the effect of rGO-FA-based nanomaterials on photothermal therapy of breast cancer cells in the microfluidic co-culture platform. From the microfluidic co-culture platform with breast cancer cells and human umbilical vein endothelial cells (HUVECs), we observed that the viability of breast cancer cells treated with rGO-FA-based functional nanomaterials was significantly decreased after near-infrared (NIR) laser irradiation. Therefore, this microfluidic co-culture platform could be a potentially powerful tool for studying cancer cell targeting and photothermal therapy.
Collapse
Affiliation(s)
- Seok Gyu Mun
- Department of Biomedical Engineering, Sogang University, Seoul, Korea
| | | | - Jong Min Lee
- Department of Mechanical Engineering, Sogang University, Seoul, Korea
| | - Jae Hyun Lim
- Department of Biomedical Engineering, Sogang University, Seoul, Korea
| | - Jang Ho Ha
- Department of Mechanical Engineering, Sogang University, Seoul, Korea
| | | | | | - Lifeng Kang
- School of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | - Bong Geun Chung
- Department of Mechanical Engineering, Sogang University, Seoul, Korea.
| |
Collapse
|
23
|
The endothelial barrier and cancer metastasis: Does the protective facet of platelet function matter? Biochem Pharmacol 2020; 176:113886. [PMID: 32113813 DOI: 10.1016/j.bcp.2020.113886] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/24/2020] [Indexed: 12/16/2022]
Abstract
Overwhelming evidence suggests that platelets have a detrimental role in promoting cancer spread via platelet-cancer cell interactions linked to thrombotic mechanisms. On the other hand, a beneficial role of platelets in the preservation of the endothelial barrier in inflammatory conditions has been recently described, a phenomenon that could also operate in cancer-related inflammation. It is tempting to speculate that some antiplatelet strategies to combat cancer metastasis may impair the endogenous platelet-dependent mechanisms preserving endothelial barrier function. If the protective function of platelets is impaired, it may lead to increased endothelial permeability and more efficient cancer cell intravasation in the primary tumor and cancer cell extravasation at metastatic sites. In this commentary, we discuss current evidence that could support this hypothesis.
Collapse
|
24
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
25
|
Puder S, Fischer T, Mierke CT. The transmembrane protein fibrocystin/polyductin regulates cell mechanics and cell motility. Phys Biol 2019; 16:066006. [PMID: 31398719 DOI: 10.1088/1478-3975/ab39fa] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Polycystic kidney disease is a disorder that leads to fluid filled cysts that replace normal renal tubes. During the process of cellular development and in the progression of the diseases, fibrocystin can lead to impaired organ formation and even cause organ defects. Besides cellular polarity, mechanical properties play major roles in providing the optimal apical-basal or anterior-posterior symmetry within epithelial cells. A breakdown of the cell symmetry that is usually associated with mechanical property changes and it is known to be essential in many biological processes such as cell migration, polarity and pattern formation especially during development and diseases such as the autosomal recessive cystic kidney disease. Since the breakdown of the cell symmetry can be evoked by several proteins including fibrocystin, we hypothesized that cell mechanics are altered by fibrocystin. However, the effect of fibrocystin on cell migration and cellular mechanical properties is still unclear. In order to explore the function of fibrocystin on cell migration and mechanics, we analyzed fibrocystin knockdown epithelial cells in comparison to fibrocystin control cells. We found that invasiveness of fibrocystin knockdown cells into dense 3D matrices was increased and more efficient compared to control cells. Using optical cell stretching and atomic force microscopy, fibrocystin knockdown cells were more deformable and exhibited weaker cell-matrix as well as cell-cell adhesion forces, respectively. In summary, these findings show that fibrocystin knockdown cells displayed increased 3D matrix invasion through providing increased cellular deformability, decreased cell-matrix and reduced cell-cell adhesion forces.
Collapse
|
26
|
Mierke CT. The Role of the Optical Stretcher Is Crucial in the Investigation of Cell Mechanics Regulating Cell Adhesion and Motility. Front Cell Dev Biol 2019; 7:184. [PMID: 31552247 PMCID: PMC6736998 DOI: 10.3389/fcell.2019.00184] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 08/21/2019] [Indexed: 12/14/2022] Open
Abstract
The mechanical properties of cells, tissues, and the surrounding extracellular matrix environment play important roles in the process of cell adhesion and migration. In physiological and pathological processes of the cells, such as wound healing and cancer, the capacity to migrate through the extracellular matrix is crucial. Hence biophysical techniques were used to determine the mechanical properties of cells that facilitate the various migratory capacities. Since the field of mechanobiology is rapidly growing, the reliable and reproducible characterization of cell mechanics is required that facilitates the adhesion and migration of cells. One of these cell mechanical techniques is the optical stretching device, which was originally developed to investigate the mechanical properties of cells, such as the deformation of single cells in suspension. After discussing the strengths and weaknesses of the technology, the latest findings in optical stretching-based cell mechanics are presented in this review. Finally, the mechanical properties of cells are correlated with their migratory potential and it is pointed out how the inhibition of biomolecules that contribute to the to the maintenance of cytoskeletal structures in cells affect their mechanical deformability.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute for Soft Matter Physics, Faculty of Physics and Earth Sciences, Leipzig University, Leipzig, Germany
| |
Collapse
|
27
|
Mierke CT. The matrix environmental and cell mechanical properties regulate cell migration and contribute to the invasive phenotype of cancer cells. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2019; 82:064602. [PMID: 30947151 DOI: 10.1088/1361-6633/ab1628] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The minimal structural unit of a solid tumor is a single cell or a cellular compartment such as the nucleus. A closer look inside the cells reveals that there are functional compartments or even structural domains determining the overall properties of a cell such as the mechanical phenotype. The mechanical interaction of these living cells leads to the complex organization such as compartments, tissues and organs of organisms including mammals. In contrast to passive non-living materials, living cells actively respond to the mechanical perturbations occurring in their microenvironment during diseases such as fibrosis and cancer. The transformation of single cancer cells in highly aggressive and hence malignant cancer cells during malignant cancer progression encompasses the basement membrane crossing, the invasion of connective tissue, the stroma microenvironments and transbarrier migration, which all require the immediate interaction of the aggressive and invasive cancer cells with the surrounding extracellular matrix environment including normal embedded neighboring cells. All these steps of the metastatic pathway seem to involve mechanical interactions between cancer cells and their microenvironment. The pathology of cancer due to a broad heterogeneity of cancer types is still not fully understood. Hence it is necessary to reveal the signaling pathways such as mechanotransduction pathways that seem to be commonly involved in the development and establishment of the metastatic and mechanical phenotype in several carcinoma cells. We still do not know whether there exist distinct metastatic genes regulating the progression of tumors. These metastatic genes may then be activated either during the progression of cancer by themselves on their migration path or in earlier stages of oncogenesis through activated oncogenes or inactivated tumor suppressor genes, both of which promote the metastatic phenotype. In more detail, the adhesion of cancer cells to their surrounding stroma induces the generation of intracellular contraction forces that deform their microenvironments by alignment of fibers. The amplitude of these forces can adapt to the mechanical properties of the microenvironment. Moreover, the adhesion strength of cancer cells seems to determine whether a cancer cell is able to migrate through connective tissue or across barriers such as the basement membrane or endothelial cell linings of blood or lymph vessels in order to metastasize. In turn, exposure of adherent cancer cells to physical forces, such as shear flow in vessels or compression forces around tumors, reinforces cell adhesion, regulates cell contractility and restructures the ordering of the local stroma matrix that leads subsequently to secretion of crosslinking proteins or matrix degrading enzymes. Hence invasive cancer cells alter the mechanical properties of their microenvironment. From a mechanobiological point-of-view, the recognized physical signals are transduced into biochemical signaling events that guide cellular responses such as cancer progression after the malignant transition of cancer cells from an epithelial and non-motile phenotype to a mesenchymal and motile (invasive) phenotype providing cellular motility. This transition can also be described as the physical attempt to relate this cancer cell transitional behavior to a T1 phase transition such as the jamming to unjamming transition. During the invasion of cancer cells, cell adaptation occurs to mechanical alterations of the local stroma, such as enhanced stroma upon fibrosis, and therefore we need to uncover underlying mechano-coupling and mechano-regulating functional processes that reinforce the invasion of cancer cells. Moreover, these mechanisms may also be responsible for the awakening of dormant residual cancer cells within the microenvironment. Physicists were initially tempted to consider the steps of the cancer metastasis cascade as single events caused by a single mechanical alteration of the overall properties of the cancer cell. However, this general and simple view has been challenged by the finding that several mechanical properties of cancer cells and their microenvironment influence each other and continuously contribute to tumor growth and cancer progression. In addition, basement membrane crossing, cell invasion and transbarrier migration during cancer progression is explained in physical terms by applying physical principles on living cells regardless of their complexity and individual differences of cancer types. As a novel approach, the impact of the individual microenvironment surrounding cancer cells is also included. Moreover, new theories and models are still needed to understand why certain cancers are malignant and aggressive, while others stay still benign. However, due to the broad variety of cancer types, there may be various pathways solely suitable for specific cancer types and distinct steps in the process of cancer progression. In this review, physical concepts and hypotheses of cancer initiation and progression including cancer cell basement membrane crossing, invasion and transbarrier migration are presented and discussed from a biophysical point-of-view. In addition, the crosstalk between cancer cells and a chronically altered microenvironment, such as fibrosis, is discussed including the basic physical concepts of fibrosis and the cellular responses to mechanical stress caused by the mechanically altered microenvironment. Here, is highlighted how biophysical approaches, both experimentally and theoretically, have an impact on classical hallmarks of cancer and fibrosis and how they contribute to the understanding of the regulation of cancer and its progression by sensing and responding to the physical environmental properties through mechanotransduction processes. Finally, this review discusses various physical models of cell migration such as blebbing, nuclear piston, protrusive force and unjamming transition migration modes and how they contribute to cancer progression. Moreover, these cellular migration modes are influenced by microenvironmental perturbances such as fibrosis that can induce mechanical alterations in cancer cells, which in turn may impact the environment. Hence, the classical hallmarks of cancer need to be refined by including biomechanical properties of cells, cell clusters and tissues and their microenvironment to understand mechano-regulatory processes within cancer cells and the entire organism.
Collapse
|
28
|
Kunschmann T, Puder S, Fischer T, Steffen A, Rottner K, Mierke CT. The Small GTPase Rac1 Increases Cell Surface Stiffness and Enhances 3D Migration Into Extracellular Matrices. Sci Rep 2019; 9:7675. [PMID: 31118438 PMCID: PMC6531482 DOI: 10.1038/s41598-019-43975-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 05/07/2019] [Indexed: 01/21/2023] Open
Abstract
Membrane ruffling and lamellipodia formation promote the motility of adherent cells in two-dimensional motility assays by mechano-sensing of the microenvironment and initiation of focal adhesions towards their surroundings. Lamellipodium formation is stimulated by small Rho GTPases of the Rac subfamily, since genetic removal of these GTPases abolishes lamellipodium assembly. The relevance of lamellipodial or invadopodial structures for facilitating cellular mechanics and 3D cell motility is still unclear. Here, we hypothesized that Rac1 affects cell mechanics and facilitates 3D invasion. Thus, we explored whether fibroblasts that are genetically deficient for Rac1 (lacking Rac2 and Rac3) harbor altered mechanical properties, such as cellular deformability, intercellular adhesion forces and force exertion, and exhibit alterations in 3D motility. Rac1 knockout and control cells were analyzed for changes in deformability by applying an external force using an optical stretcher. Five Rac1 knockout cell lines were pronouncedly more deformable than Rac1 control cells upon stress application. Using AFM, we found that cell-cell adhesion forces are increased in Rac1 knockout compared to Rac1-expressing fibroblasts. Since mechanical deformability, cell-cell adhesion strength and 3D motility may be functionally connected, we investigated whether increased deformability of Rac1 knockout cells correlates with changes in 3D motility. All five Rac1 knockout clones displayed much lower 3D motility than Rac1-expressing controls. Moreover, force exertion was reduced in Rac1 knockout cells, as assessed by 3D fiber displacement analysis. Interference with cellular stiffness through blocking of actin polymerization by Latrunculin A could not further reduce invasion of Rac1 knockout cells. In contrast, Rac1-expressing controls treated with Latrunculin A were again more deformable and less invasive, suggesting actin polymerization is a major determinant of observed Rac1-dependent effects. Together, we propose that regulation of 3D motility by Rac1 partly involves cellular mechanics such as deformability and exertion of forces.
Collapse
Affiliation(s)
- Tom Kunschmann
- University of Leipzig, Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Linnestr. 5, 04103, Leipzig, Germany
| | - Stefanie Puder
- University of Leipzig, Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Linnestr. 5, 04103, Leipzig, Germany
| | - Tony Fischer
- University of Leipzig, Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Linnestr. 5, 04103, Leipzig, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstr. 7, 38124, Braunschweig, Germany
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstr. 7, 38106, Braunschweig, Germany
| | - Claudia Tanja Mierke
- University of Leipzig, Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Linnestr. 5, 04103, Leipzig, Germany.
| |
Collapse
|
29
|
Sauer F, Oswald L, Ariza de Schellenberger A, Tzschätzsch H, Schrank F, Fischer T, Braun J, Mierke CT, Valiullin R, Sack I, Käs JA. Collagen networks determine viscoelastic properties of connective tissues yet do not hinder diffusion of the aqueous solvent. SOFT MATTER 2019; 15:3055-3064. [PMID: 30912548 DOI: 10.1039/c8sm02264j] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Collagen accounts for the major extracellular matrix (ECM) component in many tissues and provides mechanical support for cells. Magnetic Resonance (MR) Imaging, MR based diffusion measurements and MR Elastography (MRE) are considered sensitive to the microstructure of tissues including collagen networks of the ECM. However, little is known whether water diffusion interacts with viscoelastic properties of tissues. This study combines highfield MR based diffusion measurements, novel compact tabletop MRE and confocal microscopy in collagen networks of different cross-linking states (untreated collagen gels versus additional treatment with glutaraldehyde). The consistency of bulk rheology and MRE within a wide dynamic range is demonstrated in heparin gels, a viscoelastic standard for MRE. Additional crosslinking of collagen led to an 8-fold increased storage modulus, a 4-fold increased loss modulus and a significantly decreased power law exponent, describing multi-relaxational behavior, corresponding to a pronounced transition from viscous-soft to elastic-rigid properties. Collagen network changes were not detectable by MR based diffusion measurements and microscopy which are sensitive to the micrometer scale. The MRE-measured shear modulus is sensitive to collagen fiber interactions which take place on the intrafiber level such as fiber stiffness. The insensitivity of MR based diffusion measurements to collagen hydrogels of different cross-linking states alludes that congeneric collagen structures in connective tissues do not hinder extracellular diffusive water transport. Furthermore, the glutaraldehyde induced rigorous changes in viscoelastic properties indicate that intrafibrillar dissipation is the dominant mode of viscous dissipation in collagen-dominated connective tissue.
Collapse
Affiliation(s)
- Frank Sauer
- Soft Matter Physics Division, Peter Debye Institute for Soft Matter Physics, Linnestr. 5, Leipzig, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Crosstalk between cancer cells and endothelial cells: implications for tumor progression and intervention. Arch Pharm Res 2018; 41:711-724. [DOI: 10.1007/s12272-018-1051-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/26/2018] [Indexed: 02/07/2023]
|
31
|
Fischer T, Wilharm N, Hayn A, Mierke CT. Matrix and cellular mechanical properties are the driving factors for facilitating human cancer cell motility into 3D engineered matrices. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017. [DOI: 10.1088/2057-1739/aa8bbb] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
32
|
Collateral Damage Intended-Cancer-Associated Fibroblasts and Vasculature Are Potential Targets in Cancer Therapy. Int J Mol Sci 2017; 18:ijms18112355. [PMID: 29112161 PMCID: PMC5713324 DOI: 10.3390/ijms18112355] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 10/25/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023] Open
Abstract
After oncogenic transformation, tumor cells rewire their metabolism to obtain sufficient energy and biochemical building blocks for cell proliferation, even under hypoxic conditions. Glucose and glutamine become their major limiting nutritional demands. Instead of being autonomous, tumor cells change their immediate environment not only by their metabolites but also by mediators, such as juxtacrine cell contacts, chemokines and other cytokines. Thus, the tumor cells shape their microenvironment as well as induce resident cells, such as fibroblasts and endothelial cells (ECs), to support them. Fibroblasts differentiate into cancer-associated fibroblasts (CAFs), which produce a qualitatively and quantitatively different extracellular matrix (ECM). By their contractile power, they exert tensile forces onto this ECM, leading to increased intratumoral pressure. Moreover, along with enhanced cross-linkage of the ECM components, CAFs thus stiffen the ECM. Attracted by tumor cell- and CAF-secreted vascular endothelial growth factor (VEGF), ECs sprout from pre-existing blood vessels during tumor-induced angiogenesis. Tumor vessels are distinct from EC-lined vessels, because tumor cells integrate into the endothelium or even mimic and replace it in vasculogenic mimicry (VM) vessels. Not only the VM vessels but also the characteristically malformed EC-lined tumor vessels are typical for tumor tissue and may represent promising targets in cancer therapy.
Collapse
|
33
|
Oscillations of ultra-weak photon emission from cancer and non-cancer cells stressed by culture medium change and TNF-α. Sci Rep 2017; 7:11249. [PMID: 28900100 PMCID: PMC5596028 DOI: 10.1038/s41598-017-10949-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 06/29/2017] [Indexed: 12/16/2022] Open
Abstract
Cells spontaneously emit photons in the UV to visible/near-infrared range (ultra-weak photon emission, UPE). Perturbations of the cells’ state cause changes in UPE (evoked UPE). The aim of the present study was to analyze the evoked UPE dynamics of cells caused by two types of cell perturbations (stressors): (i) a cell culture medium change, and (ii) application of the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α). Four types of human cell lines were used (squamous cell carcinoma cells, A431; adenocarcinomic alveolar basal epithelial cells, A549; p53-deficient keratinocytes, HaCaT, and cervical cancer cells, HeLa). In addition to the medium change, TNF-α was applied at different concentrations (5, 10, 20, and 40 ng/mL) and UPE measurements were performed after incubation times of 0, 30, 60, 90 min, 2, 5, 12, 24, 48 h. It was observed that (i) the change of cell culture medium (without added TNF-α) induces a cell type-specific transient increase in UPE with the largest UPE increase observed in A549 cells, (ii) the addition of TNF-α induces a cell type-specific and dose-dependent change in UPE, and (iii) stressed cell cultures in general exhibit oscillatory UPE changes.
Collapse
|
34
|
A Biomimetic Microfluidic Tumor Microenvironment Platform Mimicking the EPR Effect for Rapid Screening of Drug Delivery Systems. Sci Rep 2017; 7:9359. [PMID: 28839211 PMCID: PMC5571192 DOI: 10.1038/s41598-017-09815-9] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/31/2017] [Indexed: 01/18/2023] Open
Abstract
Real-time monitoring of tumor drug delivery in vivo is a daunting challenge due to the heterogeneity and complexity of the tumor microenvironment. In this study, we developed a biomimetic microfluidic tumor microenvironment (bMTM) comprising co-culture of tumor and endothelial cells in a 3D environment. The platform consists of a vascular compartment featuring a network of vessels cultured with endothelial cells forming a complete lumen under shear flow in communication with 3D solid tumors cultured in a tumor compartment. Endothelial cell permeability to both small dye molecules and large liposomal drug carriers were quantified using fluorescence microscopy. Endothelial cell intercellular junction formation was characterized by immunostaining. Endothelial cell permeability significantly increased in the presence of either tumor cell conditioned media (TCM) or tumor cells. The magnitude of this increase in permeability was significantly higher in the presence of metastatic breast tumor cells as compared to non-metastatic ones. Immunostaining revealed impaired endothelial cell-cell junctions in the presence of either metastatic TCM or metastatic tumor cells. Our findings indicate that the bMTM platform mimics the tumor microenvironment including the EPR effect. This platform has a significant potential in applications such as cell-cell/cell-drug carrier interaction studies and rapid screening of cancer drug therapeutics/carriers.
Collapse
|
35
|
Haffner MC, Esopi DM, Chaux A, Gürel M, Ghosh S, Vaghasia AM, Tsai H, Kim K, Castagna N, Lam H, Hicks J, Wyhs N, Biswal Shinohara D, Hurley PJ, Simons BW, Schaeffer EM, Lotan TL, Isaacs WB, Netto GJ, De Marzo AM, Nelson WG, An SS, Yegnasubramanian S. AIM1 is an actin-binding protein that suppresses cell migration and micrometastatic dissemination. Nat Commun 2017; 8:142. [PMID: 28747635 PMCID: PMC5529512 DOI: 10.1038/s41467-017-00084-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 06/01/2017] [Indexed: 01/05/2023] Open
Abstract
A defining hallmark of primary and metastatic cancers is the migration and invasion of malignant cells. These invasive properties involve altered dynamics of the cytoskeleton and one of its major structural components β-actin. Here we identify AIM1 (absent in melanoma 1) as an actin-binding protein that suppresses pro-invasive properties in benign prostate epithelium. Depletion of AIM1 in prostate epithelial cells increases cytoskeletal remodeling, intracellular traction forces, cell migration and invasion, and anchorage-independent growth. In addition, decreased AIM1 expression results in increased metastatic dissemination in vivo. AIM1 strongly associates with the actin cytoskeleton in prostate epithelial cells in normal tissues, but not in prostate cancers. In addition to a mislocalization of AIM1 from the actin cytoskeleton in invasive cancers, advanced prostate cancers often harbor AIM1 deletion and reduced expression. These findings implicate AIM1 as a key suppressor of invasive phenotypes that becomes dysregulated in primary and metastatic prostate cancer. Invasion of malignant cells involves changes in cytoskeleton dynamics. Here the authors identify absent in melanoma 1 as an actin binding protein and show that it regulates cytoskeletal remodeling and cell migration in prostate epithelial cells, acting as a metastatic suppressor in cancer cells.
Collapse
Affiliation(s)
- Michael C Haffner
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - David M Esopi
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Alcides Chaux
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Office of Scientific Research, Norte University, Asunción,, Paraguay
| | - Meltem Gürel
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Susmita Ghosh
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Ajay M Vaghasia
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Harrison Tsai
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Kunhwa Kim
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Nicole Castagna
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Hong Lam
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Jessica Hicks
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Nicolas Wyhs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | | | - Paula J Hurley
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Brian W Simons
- Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Edward M Schaeffer
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Tamara L Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - William B Isaacs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - George J Netto
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Angelo M De Marzo
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - William G Nelson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Steven S An
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.,Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.,Johns Hopkins Physical Sciences in Oncology Center, Johns Hopkins University, Baltimore, MD, USA
| | - Srinivasan Yegnasubramanian
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA. .,Department of Pathology, Johns Hopkins University, Baltimore, MD, USA. .,Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA. .,Brady Urological Institute, School of Medicine, Johns Hopkins University, Baltimore, MD, USA. .,Johns Hopkins Physical Sciences in Oncology Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
36
|
Mierke CT, Fischer T, Puder S, Kunschmann T, Soetje B, Ziegler WH. Focal adhesion kinase activity is required for actomyosin contractility-based invasion of cells into dense 3D matrices. Sci Rep 2017; 7:42780. [PMID: 28202937 PMCID: PMC5311912 DOI: 10.1038/srep42780] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 01/16/2017] [Indexed: 02/07/2023] Open
Abstract
The focal adhesion kinase (FAK) regulates the dynamics of integrin-based cell adhesions important for motility. FAK's activity regulation is involved in stress-sensing and focal-adhesion turnover. The effect of FAK on 3D migration and cellular mechanics is unclear. We analyzed FAK knock-out mouse embryonic fibroblasts and cells expressing a kinase-dead FAK mutant, R454-FAK, in comparison to FAK wild-type cells. FAK knock-out and FAKR454/R454 cells invade dense 3D matrices less efficiently. These results are supported by FAK knock-down in wild-type fibroblasts and MDA-MB-231 human breast cancer cells showing reduced invasiveness. Pharmacological interventions indicate that in 3D matrices, cells deficient in FAK or kinase-activity behave similarly to wild-type cells treated with inhibitors of Src-activity or actomyosin-contractility. Using magnetic tweezers experiments, FAKR454/R454 cells are shown to be softer and exhibit impaired adhesion to fibronectin and collagen, which is consistent with their reduced 3D invasiveness. In line with this, FAKR454/R454 cells cannot contract the matrix in contrast to FAK wild-type cells. Finally, our findings demonstrate that active FAK facilitates 3D matrix invasion through increased cellular stiffness and transmission of actomyosin-dependent contractile force in dense 3D extracellular matrices.
Collapse
Affiliation(s)
- Claudia T. Mierke
- Institute of Experimental Physics I, Biological Physics Division, Faculty of Physics and Earth Science, University of Leipzig, Leipzig, Germany
| | - Tony Fischer
- Institute of Experimental Physics I, Biological Physics Division, Faculty of Physics and Earth Science, University of Leipzig, Leipzig, Germany
| | - Stefanie Puder
- Institute of Experimental Physics I, Biological Physics Division, Faculty of Physics and Earth Science, University of Leipzig, Leipzig, Germany
| | - Tom Kunschmann
- Institute of Experimental Physics I, Biological Physics Division, Faculty of Physics and Earth Science, University of Leipzig, Leipzig, Germany
| | - Birga Soetje
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Wolfgang H. Ziegler
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany
| |
Collapse
|
37
|
Wojtukiewicz MZ, Hempel D, Sierko E, Tucker SC, Honn KV. Thrombin-unique coagulation system protein with multifaceted impacts on cancer and metastasis. Cancer Metastasis Rev 2017; 35:213-33. [PMID: 27189210 DOI: 10.1007/s10555-016-9626-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The association between blood coagulation and cancer development is well recognized. Thrombin, the pleiotropic enzyme best known for its contribution to fibrin formation and platelet aggregation during vascular hemostasis, may also trigger cellular events through protease-activated receptors, PAR-1 and PAR-4, leading to cancer progression. Our pioneering findings provided evidence that thrombin contributes to cancer metastasis by increasing adhesive potential of malignant cells. However, there is evidence that thrombin regulates every step of cancer dissemination: (1) cancer cell invasion, detachment from primary tumor, migration; (2) entering the blood vessel; (3) surviving in vasculature; (4) extravasation; (5) implantation in host organs. Recent studies have provided new molecular data about thrombin generation in cancer patients and the mechanisms by which thrombin contributes to transendothelial migration, platelet/tumor cell interactions, angiogenesis, and other processes. Though a great deal is known regarding the role of thrombin in cancer dissemination, there are new data for multiple thrombin-mediated events that justify devoting focus to this topic with a comprehensive approach.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland. .,Department of Clinical Oncology, Comprehensive Cancer Center in Bialystok, Bialystok, Poland.
| | - Dominika Hempel
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland.,Department of Radiotherapy, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Ewa Sierko
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland.,Department of Radiotherapy, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Stephanie C Tucker
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Wayne State University, Detroit, MI, USA
| | - Kenneth V Honn
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Wayne State University, Detroit, MI, USA.,Department of Chemistry, Wayne State University, Detroit, MI, USA.,Department of Oncology, Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
38
|
Tanja Mierke C. Physical role of nuclear and cytoskeletal confinements in cell migration mode selection and switching. AIMS BIOPHYSICS 2017. [DOI: 10.3934/biophy.2017.4.615] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
39
|
Kunschmann T, Puder S, Fischer T, Perez J, Wilharm N, Mierke CT. Integrin-linked kinase regulates cellular mechanics facilitating the motility in 3D extracellular matrices. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:580-593. [PMID: 28011283 DOI: 10.1016/j.bbamcr.2016.12.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022]
Abstract
The motility of cells plays an important role for many processes such as wound healing and malignant progression of cancer. The efficiency of cell motility is affected by the microenvironment. The connection between the cell and its microenvironment is facilitated by cell-matrix adhesion receptors and upon their activation focal adhesion proteins such as integrin-linked kinase (ILK) are recruited to sites of focal adhesion formation. In particular, ILK connects cell-matrix receptors to the actomyosin cytoskeleton. However, ILK's role in cell mechanics regulating cellular motility in 3D collagen matrices is still not well understood. We suggest that ILK facilitates 3D motility by regulating cellular mechanical properties such as stiffness and force transmission. Thus, ILK wild-type and knock-out cells are analyzed for their ability to migrate on 2D substrates serving as control and in dense 3D extracellular matrices. Indeed, ILK wild-type cells migrated faster on 2D substrates and migrated more numerous and deeper in 3D matrices. Hence, we analyzed cellular deformability, Young's modulus (stiffness) and adhesion forces. We found that ILK wild-type cells are less deformable (stiffer) and produce higher cell-matrix adhesion forces compared to ILK knock-out cells. Finally, ILK is essential for providing cellular mechanical stiffness regulating 3D motility.
Collapse
Affiliation(s)
- Tom Kunschmann
- University of Leipzig, Faculty of Physics and Earth Science, Institute for Experimental Physics I, Biological Physics Division, Linnestrasse 5, 04103 Leipzig, Germany
| | - Stefanie Puder
- University of Leipzig, Faculty of Physics and Earth Science, Institute for Experimental Physics I, Biological Physics Division, Linnestrasse 5, 04103 Leipzig, Germany
| | - Tony Fischer
- University of Leipzig, Faculty of Physics and Earth Science, Institute for Experimental Physics I, Biological Physics Division, Linnestrasse 5, 04103 Leipzig, Germany
| | - Jeremy Perez
- University of Leipzig, Faculty of Physics and Earth Science, Institute for Experimental Physics I, Biological Physics Division, Linnestrasse 5, 04103 Leipzig, Germany
| | - Nils Wilharm
- University of Leipzig, Faculty of Physics and Earth Science, Institute for Experimental Physics I, Biological Physics Division, Linnestrasse 5, 04103 Leipzig, Germany
| | - Claudia Tanja Mierke
- University of Leipzig, Faculty of Physics and Earth Science, Institute for Experimental Physics I, Biological Physics Division, Linnestrasse 5, 04103 Leipzig, Germany.
| |
Collapse
|
40
|
Abstract
Cellular motility is essential for many processes such as embryonic development, wound healing processes, tissue assembly and regeneration, immune cell trafficing and diseases such as cancer. The migration efficiency and the migratory potential depend on the type of migration mode. The previously established migration modes such as epithelial (non-migratory) and mesenchymal (migratory) as well as amoeboid (squeezing motility) relay mainly on phenomenological criteria such as cell morphology and molecular biological criteria such as gene expression. However, the physical view on the migration modes is still not well understood. As the process of malignant cancer progression such as metastasis depends on the migration of single cancer cells and their migration mode, this review focuses on the different migration strategies and discusses which mechanical prerequisites are necessary to perform a special migration mode through a 3-dimensional microenvironment. In particular, this review discusses how cells can distinguish and finally switch between the migration modes and what impact do the physical properties of cells and their microenvironment have on the transition between the novel migration modes such as blebbing and protrusive motility.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- a Faculty of Physics and Earth Science; Institute of Experimental Physics I; Biological Physics Division; University of Leipzig ; Leipzig , Germany
| |
Collapse
|
41
|
Williams KC, Wong E, Leong HS, Jackson DN, Allan AL, Chambers AF. Cancer dissemination from a physical sciences perspective. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2016. [DOI: 10.1088/2057-1739/2/2/023001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
42
|
Lautscham LA, Kämmerer C, Lange JR, Kolb T, Mark C, Schilling A, Strissel PL, Strick R, Gluth C, Rowat AC, Metzner C, Fabry B. Migration in Confined 3D Environments Is Determined by a Combination of Adhesiveness, Nuclear Volume, Contractility, and Cell Stiffness. Biophys J 2016; 109:900-13. [PMID: 26331248 DOI: 10.1016/j.bpj.2015.07.025] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/13/2023] Open
Abstract
In cancer metastasis and other physiological processes, cells migrate through the three-dimensional (3D) extracellular matrix of connective tissue and must overcome the steric hindrance posed by pores that are smaller than the cells. It is currently assumed that low cell stiffness promotes cell migration through confined spaces, but other factors such as adhesion and traction forces may be equally important. To study 3D migration under confinement in a stiff (1.77 MPa) environment, we use soft lithography to fabricate polydimethylsiloxane (PDMS) devices consisting of linear channel segments with 20 μm length, 3.7 μm height, and a decreasing width from 11.2 to 1.7 μm. To study 3D migration in a soft (550 Pa) environment, we use self-assembled collagen networks with an average pore size of 3 μm. We then measure the ability of four different cancer cell lines to migrate through these 3D matrices, and correlate the results with cell physical properties including contractility, adhesiveness, cell stiffness, and nuclear volume. Furthermore, we alter cell adhesion by coating the channel walls with different amounts of adhesion proteins, and we increase cell stiffness by overexpression of the nuclear envelope protein lamin A. Although all cell lines are able to migrate through the smallest 1.7 μm channels, we find significant differences in the migration velocity. Cell migration is impeded in cell lines with larger nuclei, lower adhesiveness, and to a lesser degree also in cells with lower contractility and higher stiffness. Our data show that the ability to overcome the steric hindrance of the matrix cannot be attributed to a single cell property but instead arises from a combination of adhesiveness, nuclear volume, contractility, and cell stiffness.
Collapse
Affiliation(s)
- Lena A Lautscham
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany.
| | - Christoph Kämmerer
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Janina R Lange
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Thorsten Kolb
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Christoph Mark
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Achim Schilling
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Pamela L Strissel
- Laboratory for Molecular Medicine, Department of Gynecology and Obstetrics, University-Clinic Erlangen, Erlangen, Germany
| | - Reiner Strick
- Laboratory for Molecular Medicine, Department of Gynecology and Obstetrics, University-Clinic Erlangen, Erlangen, Germany
| | - Caroline Gluth
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Amy C Rowat
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, California
| | - Claus Metzner
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ben Fabry
- Biophysics Group, Department of Physics, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
43
|
Connor Y, Tekleab S, Nandakumar S, Walls C, Tekleab Y, Husain A, Gadish O, Sabbisetti V, Kaushik S, Sehrawat S, Kulkarni A, Dvorak H, Zetter B, R Edelman E, Sengupta S. Physical nanoscale conduit-mediated communication between tumour cells and the endothelium modulates endothelial phenotype. Nat Commun 2015; 6:8671. [PMID: 26669454 PMCID: PMC4697439 DOI: 10.1038/ncomms9671] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 09/18/2015] [Indexed: 02/06/2023] Open
Abstract
Metastasis is a major cause of mortality and remains a hurdle in the search for a cure for cancer. Not much is known about metastatic cancer cells and endothelial cross-talk, which occurs at multiple stages during metastasis. Here we report a dynamic regulation of the endothelium by cancer cells through the formation of nanoscale intercellular membrane bridges, which act as physical conduits for transfer of microRNAs. The communication between the tumour cell and the endothelium upregulates markers associated with pathological endothelium, which is reversed by pharmacological inhibition of these nanoscale conduits. These results lead us to define the notion of ‘metastatic hijack': cancer cell-induced transformation of healthy endothelium into pathological endothelium via horizontal communication through the nanoscale conduits. Pharmacological perturbation of these nanoscale membrane bridges decreases metastatic foci in vivo. Targeting these nanoscale membrane bridges may potentially emerge as a new therapeutic opportunity in the management of metastatic cancer. Cancer cells and stromal cells have been shown to pass cellular information between each other via exosomes. Here, the authors demonstrate that cancer cells can communicate with endothelial cells through nanoscale membrane bridges, and demonstrate that microRNAs are passed through these nanobridges, which modulates endothelial cell phenotype.
Collapse
Affiliation(s)
- Yamicia Connor
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts 02139, USA.,Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.,Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Sarah Tekleab
- Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA
| | - Shyama Nandakumar
- Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA
| | - Cherelle Walls
- Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA
| | - Yonatan Tekleab
- Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Amjad Husain
- Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Or Gadish
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts 02139, USA.,Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Venkata Sabbisetti
- Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Shelly Kaushik
- India Innovation Research Center, New Delhi 110092, India
| | - Seema Sehrawat
- Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA
| | - Ashish Kulkarni
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts 02139, USA.,Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Harold Dvorak
- Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | - Bruce Zetter
- Harvard Medical School, Boston, Massachusetts 02115, USA.,Program in Vascular Biology and Department of Surgery, Children's Hospital, Boston, Massachusetts 02115, USA.,Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | - Elazer R Edelman
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts 02139, USA.,Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.,Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Shiladitya Sengupta
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts 02139, USA.,Department of Medicine, Brigham and Women's Hospital, 65 Landsdowne Street, Room 317, Boston, Massachusetts 02115, USA.,Harvard Medical School, Boston, Massachusetts 02115, USA.,Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA
| |
Collapse
|
44
|
Rubtsova SN, Zhitnyak IY, Gloushankova NA. A Novel Role of E-Cadherin-Based Adherens Junctions in Neoplastic Cell Dissemination. PLoS One 2015; 10:e0133578. [PMID: 26207916 PMCID: PMC4514802 DOI: 10.1371/journal.pone.0133578] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/29/2015] [Indexed: 12/12/2022] Open
Abstract
Using confocal microscopy, we analyzed the behavior of IAR-6-1, IAR1170, and IAR1162 transformed epithelial cells seeded onto the confluent monolayer of normal IAR-2 epithelial cells. Live-cell imaging of neoplastic cells stably expressing EGFP and of normal epithelial cells stably expressing mKate2 showed that transformed cells retaining expression of E-cadherin were able to migrate over the IAR-2 epithelial monolayer and invade the monolayer. Transformed IAR cells invaded the IAR-2 monolayer at the boundaries between normal cells. Studying interactions of IAR-6-1 transformed cells stably expressing GFP-E-cadherin with the IAR-2 epithelial monolayer, we found that IAR-6-1 cells established E-cadherin-based adhesions with normal epithelial cells: dot-like dynamic E-cadherin-based adhesions in protrusions and large adherens junctions at the cell sides and rear. A comparative study of a panel of transformed IAR cells that differ by their ability to form E-cadherin-based AJs, either through loss of E-cadherin expression or through expression of a dominant negative E-cadherin mutant, demonstrated that E-cadherin-based AJs are key mediators of the interactions between neoplastic and normal epithelial cells. IAR-6-1DNE cells expressing a dominant-negative mutant form of E-cadherin with the mutation in the first extracellular domain practically lost the ability to adhere to IAR-2 cells and invade the IAR-2 epithelial monolayer. The ability of cancer cells to form E-cadherin-based AJs with the surrounding normal epithelial cells may play an important role in driving cancer cell dissemination in the body.
Collapse
Affiliation(s)
- Svetlana N. Rubtsova
- Institute of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | - Irina Y. Zhitnyak
- Institute of Carcinogenesis, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia
| | | |
Collapse
|
45
|
Hamilla SM, Stroka KM, Aranda-Espinoza H. VE-cadherin-independent cancer cell incorporation into the vascular endothelium precedes transmigration. PLoS One 2014; 9:e109748. [PMID: 25275457 PMCID: PMC4183660 DOI: 10.1371/journal.pone.0109748] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/10/2014] [Indexed: 12/31/2022] Open
Abstract
Metastasis is accountable for 90% of cancer deaths. During metastasis, tumor cells break away from the primary tumor, enter the blood and the lymph vessels, and use them as highways to travel to distant sites in the body to form secondary tumors. Cancer cell migration through the endothelium and into the basement membrane represents a critical step in the metastatic cascade, yet it is not well understood. This process is well characterized for immune cells that routinely transmigrate through the endothelium to sites of infection, inflammation, or injury. Previous studies with leukocytes have demonstrated that this step depends heavily on the activation status of the endothelium and subendothelial substrate stiffness. Here, we used a previously established in vitro model of the endothelium and live cell imaging, in order to observe cancer cell transmigration and compare this process to leukocytes. Interestingly, cancer cell transmigration includes an additional step, which we term ‘incorporation’, into the endothelial cell (EC) monolayer. During this phase, cancer cells physically displace ECs, leading to the dislocation of EC VE-cadherin away from EC junctions bordering cancer cells, and spread into the monolayer. In some cases, ECs completely detach from the matrix. Furthermore, cancer cell incorporation occurs independently of the activation status and the subendothelial substrate stiffness for breast cancer and melanoma cells, a notable difference from the process by which leukocytes transmigrate. Meanwhile, pancreatic cancer cell incorporation was dependent on the activation status of the endothelium and changed on very stiff subendothelial substrates. Collectively, our results provide mechanistic insights into tumor cell extravasation and demonstrate that incorporation is one of the earliest steps.
Collapse
Affiliation(s)
- Susan M. Hamilla
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
| | - Kimberly M. Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
| | - Helim Aranda-Espinoza
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Weidert E, Pohler SE, Gomez EW, Dong C. Actinomyosin contraction, phosphorylation of VE-cadherin, and actin remodeling enable melanoma-induced endothelial cell-cell junction disassembly. PLoS One 2014; 9:e108092. [PMID: 25225982 PMCID: PMC4167543 DOI: 10.1371/journal.pone.0108092] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 08/25/2014] [Indexed: 02/07/2023] Open
Abstract
During melanoma cell extravasation through the vascular endothelium, melanoma cells interact with endothelial cells through secretion of cytokines and by adhesion between proteins displayed on opposing cell surfaces. How these tumor cell associated signals together regulate the dynamics of intracellular signaling pathways within endothelial cells leading to endothelial cell-cell junction disruption is not well understood. Here, we used a combination of experimental and computational approaches to examine the individual and combined effects of activation of the vascular cell adhesion molecule (VCAM)-1, interleukin (IL)-8, and IL-1β signaling pathways on the integrity of vascular junctions. Our simulations predict a multifaceted interplay of signaling resulting from individual activation of VCAM-1, IL-8 and IL-1β pathways that is neither synergistic nor additive compared to all inputs turned on simultaneously. Furthermore, we show that the levels of phosphorylated proteins associated with actinomyosin contractility and junction disassembly peak prior to those related to actin remodeling. The results of this work provide insight into the dynamics of tumor-mediated endothelial junction disassembly and suggest that targeting proteins downstream of several interaction pathways may be the most effective therapeutic approach to reduce melanoma extravasation.
Collapse
Affiliation(s)
- Eric Weidert
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Steven E. Pohler
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Esther W. Gomez
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (EWG); (CD)
| | - Cheng Dong
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail: (EWG); (CD)
| |
Collapse
|
47
|
Mierke CT. The fundamental role of mechanical properties in the progression of cancer disease and inflammation. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2014; 77:076602. [PMID: 25006689 DOI: 10.1088/0034-4885/77/7/076602] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The role of mechanical properties in cancer disease and inflammation is still underinvestigated and even ignored in many oncological and immunological reviews. In particular, eight classical hallmarks of cancer have been proposed, but they still ignore the mechanics behind the processes that facilitate cancer progression. To define the malignant transformation of neoplasms and finally reveal the functional pathway that enables cancer cells to promote cancer progression, these classical hallmarks of cancer require the inclusion of specific mechanical properties of cancer cells and their microenvironment such as the extracellular matrix as well as embedded cells such as fibroblasts, macrophages or endothelial cells. Thus, this review will present current cancer research from a biophysical point of view and will therefore focus on novel physical aspects and biophysical methods to investigate the aggressiveness of cancer cells and the process of inflammation. As cancer or immune cells are embedded in a certain microenvironment such as the extracellular matrix, the mechanical properties of this microenvironment cannot be neglected, and alterations of the microenvironment may have an impact on the mechanical properties of the cancer or immune cells. Here, it is highlighted how biophysical approaches, both experimental and theoretical, have an impact on the classical hallmarks of cancer and inflammation. It is even pointed out how these biophysical approaches contribute to the understanding of the regulation of cancer disease and inflammatory responses after tissue injury through physical microenvironmental property sensing mechanisms. The recognized physical signals are transduced into biochemical signaling events that guide cellular responses, such as malignant tumor progression, after the transition of cancer cells from an epithelial to a mesenchymal phenotype or an inflammatory response due to tissue injury. Moreover, cell adaptation to mechanical alterations, in particular the understanding of mechano-coupling and mechano-regulating functions in cell invasion, appears as an important step in cancer progression and inflammatory response to injuries. This may lead to novel insights into cancer disease and inflammatory diseases and will overcome classical views on cancer and inflammation. In addition, this review will discuss how the physics of cancer and inflammation can help to reveal whether cancer cells will invade connective tissue and metastasize or how leukocytes extravasate and migrate through the tissue. In this review, the physical concepts of cancer progression, including the tissue basement membrane a cancer cell is crossing, its invasion and transendothelial migration as well as the basic physical concepts of inflammatory processes and the cellular responses to the mechanical stress of the microenvironment such as external forces and matrix stiffness, are presented and discussed. In conclusion, this review will finally show how physical measurements can improve classical approaches that investigate cancer and inflammatory diseases, and how these physical insights can be integrated into classical tumor biological approaches.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Institute of Experimental Physics I, Biological Physics Division, University of Leipzig, Linnéstr. 5, 04103 Leipzig, Germany
| |
Collapse
|
48
|
Mierke CT. Phagocytized beads reduce the α5β1 integrin facilitated invasiveness of cancer cells by regulating cellular stiffness. Cell Biochem Biophys 2014; 66:599-622. [PMID: 23329175 DOI: 10.1007/s12013-012-9506-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cell invasion through the extracellular matrix (ECM) of connective tissue is an important biomechanical process, which plays a prominent role in tumor progression. The malignancy of tumors depends mainly on the capacity of cancer cells to migrate and metastasize. A prerequisite for metastasis is the invasion of cancer cells through connective tissue to targeted organs. Cellular stiffness and cytoskeletal remodeling dynamics have been proposed to affect the invasiveness of cancer cells. Here, this study investigated whether highly invasive cancer cells are capable of invading into dense 3D-ECMs with an average pore-size of 1.3 or 3.0 μm when phagocytized beads (2.7 and 4.5 μm diameter) increased their cellular stiffness and reduced their cytoskeletal remodeling dynamics compared to weakly invasive cancer cells. The phagocytized beads decreased the invasiveness of the α5β1(high) cancer cells into 3D-ECMs, whereas the invasiveness of the α5β1(low) cancer cells was not affected. The effect of phagocytized beads on the highly invasive α5β1(high) cells was abolished by specific knock-down of the α5 integrin subunit or addition of an anti-α5 integrin blocking antibody. Furthermore, the reduction of contractile forces using MLCK and ROCK inhibitors abolished the effect of phagocytized beads on the invasiveness of α5β1(high) cells. In addition, the cellular stiffness of α5β1(high) cells was increased after bead phagocytosis, whereas the bead phagocytosis did not alter the stiffness of α5β1(low) cells. Taken together, the α5β1 integrin dependent invasiveness was reduced after bead phagocytosis by altered biomechanical properties, suggesting that the α5β1(high) cells need an appropriate intermediate cellular stiffness to overcome the steric hindrance of 3D-ECMs, whereas the α5β1(low) cells were not affected by phagocytized beads.
Collapse
Affiliation(s)
- Claudia T Mierke
- Biological Physics Division, Institute of Experimental Physics I, Faculty of Physics and Earth Science, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
49
|
Gialeli C, Viola M, Barbouri D, Kletsas D, Passi A, Karamanos NK. Dynamic interplay between breast cancer cells and normal endothelium mediates the expression of matrix macromolecules, proteasome activity and functional properties of endothelial cells. Biochim Biophys Acta Gen Subj 2014; 1840:2549-59. [PMID: 24582970 DOI: 10.1016/j.bbagen.2014.02.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 02/06/2014] [Accepted: 02/14/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Breast cancer-endothelium interactions provide regulatory signals facilitating tumor progression. The endothelial cells have so far been mainly viewed in the context of tumor perfusion and relatively little is known regarding the effects of such paracrine interactions on the expression of extracellular matrix (ECM), proteasome activity and properties of endothelial cells. METHODS To address the effects of breast cancer cell (BCC) lines MDA-MB-231 and MCF-7 on the endothelial cells, two cell culture models were utilized; one involves endothelial cell culture in the presence of BCCs-derived conditioned media (CM) and the other co-culture of both cell populations in a Transwell system. Real-time PCR was utilized to evaluate gene expression, an immunofluorescence assay for proteasome activity, and functional assays (migration, adhesion and invasion) and immunofluorescence microscopy for cell integrity and properties. RESULTS BCC-CM decreases the cell migration of HUVEC. Adhesion and invasion of BCCs are favored by HUVEC and HUVEC-CM. HA levels and the expression of CD44 and HA synthase-2 by HUVEC are substantially upregulated in both cell culture approaches. Adhesion molecules, ICAM-1 and VCAM-1, are also highly upregulated, whereas MT1-MMP and MMP-2 expressions are significantly downregulated in both culture systems. Notably, the expression and activity of the proteasome β5 subunit are increased, especially by the action of MDA-MB-231-CM on HUVEC. CONCLUSIONS AND GENERAL SIGNIFICANCE BCCs significantly alter the expression of matrix macromolecules, proteasome activity and functional properties of endothelial cells. Deep understanding of such paracrine interactions will help to design novel drugs targeting breast cancer at the ECM level. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Ch Gialeli
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; Foundation for Research and Technology, Institute of Chemical Engineering Sciences (FORTH/ICE-HT), 26500 Patras, Greece
| | - M Viola
- Department of Surgery and Morphological Sciences, University of Insubria, Varese, Italy
| | - D Barbouri
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; Foundation for Research and Technology, Institute of Chemical Engineering Sciences (FORTH/ICE-HT), 26500 Patras, Greece
| | - D Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biology, National Center of Scientific Research "Demokritos", Athens, Greece
| | - A Passi
- Department of Surgery and Morphological Sciences, University of Insubria, Varese, Italy
| | - N K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Res. Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26110 Patras, Greece; Foundation for Research and Technology, Institute of Chemical Engineering Sciences (FORTH/ICE-HT), 26500 Patras, Greece.
| |
Collapse
|
50
|
Stroka KM, Konstantopoulos K. Physical biology in cancer. 4. Physical cues guide tumor cell adhesion and migration. Am J Physiol Cell Physiol 2014; 306:C98-C109. [PMID: 24133064 PMCID: PMC3919991 DOI: 10.1152/ajpcell.00289.2013] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 10/15/2013] [Indexed: 12/13/2022]
Abstract
As tumor cells metastasize from the primary tumor location to a distant secondary site, they encounter an array of biologically and physically heterogeneous microenvironments. While it is well established that biochemical signals guide all stages of the metastatic cascade, mounting evidence indicates that physical cues also direct tumor cell behavior, including adhesion and migration phenotypes. Physical cues acting on tumor cells in vivo include extracellular matrix mechanical properties, dimensionality, and topography, as well as interstitial flow, hydrodynamic shear stresses, and local forces due to neighboring cells. State-of-the-art technologies have recently enabled us and other researchers to engineer cell microenvironments that mimic specific physical properties of the cellular milieu. Through integration of these engineering strategies, along with physics, molecular biology, and imaging techniques, we have acquired new insights into tumor cell adhesion and migration mechanisms. In this review, we focus on the extravasation and invasion stages of the metastatic cascade. We first discuss the physical role of the endothelium during tumor cell extravasation and invasion and how contractility of endothelial and tumor cells contributes to the ability of tumor cells to exit the vasculature. Next, we examine how matrix dimensionality and stiffness coregulate tumor cell adhesion and migration beyond the vasculature. Finally, we summarize how tumor cells translate and respond to physical cues through mechanotransduction. Because of the critical role of tumor cell mechanotransduction at various stages of the metastatic cascade, targeting signaling pathways involved in tumor cell mechanosensing of physical stimuli may prove to be an effective therapeutic strategy for cancer patients.
Collapse
Affiliation(s)
- Kimberly M Stroka
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, Maryland
| | | |
Collapse
|