151
|
Abdollahiyan P, Oroojalian F, Baradaran B, de la Guardia M, Mokhtarzadeh A. Advanced mechanotherapy: Biotensegrity for governing metastatic tumor cell fate via modulating the extracellular matrix. J Control Release 2021; 335:596-618. [PMID: 34097925 DOI: 10.1016/j.jconrel.2021.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022]
Abstract
Mechano-transduction is the procedure of mechanical stimulus translation via cells, among substrate shear flow, topography, and stiffness into a biochemical answer. TAZ and YAP are transcriptional coactivators which are recognized as relay proteins that promote mechano-transduction within the Hippo pathway. With regard to healthy cells in homeostasis, mechano-transduction regularly restricts proliferation, and TAZ and YAP are totally inactive. During cancer development a YAP/TAZ - stimulating positive response loop is formed between the growing tumor and the stiffening ECM. As tumor developments, local stromal and cancerous cells take advantage of mechanotransduction to enhance proliferation, induce their migratory into remote tissues, and promote chemotherapeutic resistance. As a newly progresses paradigm, nanoparticle-conjunctions (such as magnetic nanoparticles, and graphene derivatives nanoparticles) hold significant promises for remote regulation of cells and their relevant events at molecular scale. Despite outstanding developments in employing nanoparticles for drug targeting studies, the role of nanoparticles on cellular behaviors (proliferation, migration, and differentiation) has still required more evaluations in the field of mechanotherapy. In this paper, the in-depth contribution of mechano-transduction is discussed during tumor progression, and how these consequences can be evaluated in vitro.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, 46100 Burjassot, Valencia, Spain
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
152
|
Di-Luoffo M, Ben-Meriem Z, Lefebvre P, Delarue M, Guillermet-Guibert J. PI3K functions as a hub in mechanotransduction. Trends Biochem Sci 2021; 46:878-888. [PMID: 34112586 DOI: 10.1016/j.tibs.2021.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/22/2021] [Accepted: 05/12/2021] [Indexed: 01/18/2023]
Abstract
Mammalian cells integrate different types of stimuli that govern their fate. These stimuli encompass biochemical as well as biomechanical cues (shear, tensile, and compressive stresses) that are usually studied separately. The phosphatidylinositol 3-kinase (PI3K) enzymes, producing signaling phosphoinositides at plasma and intracellular membranes, are key in intracellular signaling and vesicular trafficking pathways. Recent evidence in cancer research demonstrates that these enzymes are essential in mechanotransduction. Despite this, the importance of the integration of biomechanical cues and PI3K-driven biochemical signals is underestimated. In this opinion article, we make the hypothesis that modeling of biomechanical cues is critical to understand PI3K oncogenicity. We also identify known/missing knowledge in terms of isoform specificity and molecular pathways of activation, knowledge that is needed for clinical applications.
Collapse
Affiliation(s)
- M Di-Luoffo
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (Inserm) U1037, Centre National de la Recherche Scientifique (CNRS) U5071, Toulouse, France; Laboratoire D'analyse et D'architectures Des Systems (LAAS)-CNRS (Centre National de la Recherche Scientifique), Toulouse, France
| | - Z Ben-Meriem
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (Inserm) U1037, Centre National de la Recherche Scientifique (CNRS) U5071, Toulouse, France; Laboratoire D'analyse et D'architectures Des Systems (LAAS)-CNRS (Centre National de la Recherche Scientifique), Toulouse, France
| | - P Lefebvre
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (Inserm) U1037, Centre National de la Recherche Scientifique (CNRS) U5071, Toulouse, France; Laboratoire D'analyse et D'architectures Des Systems (LAAS)-CNRS (Centre National de la Recherche Scientifique), Toulouse, France
| | - M Delarue
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (Inserm) U1037, Centre National de la Recherche Scientifique (CNRS) U5071, Toulouse, France; Laboratoire D'analyse et D'architectures Des Systems (LAAS)-CNRS (Centre National de la Recherche Scientifique), Toulouse, France
| | - J Guillermet-Guibert
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Université de Toulouse, Institut National de la Santé et de la Recherche Médicale (Inserm) U1037, Centre National de la Recherche Scientifique (CNRS) U5071, Toulouse, France; TouCAN (Laboratoire d'Excellence Toulouse Cancer), Toulouse, France.
| |
Collapse
|
153
|
Vasilaki D, Bakopoulou A, Tsouknidas A, Johnstone E, Michalakis K. Biophysical interactions between components of the tumor microenvironment promote metastasis. Biophys Rev 2021; 13:339-357. [PMID: 34168685 PMCID: PMC8214652 DOI: 10.1007/s12551-021-00811-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
During metastasis, tumor cells need to adapt to their dynamic microenvironment and modify their mechanical properties in response to both chemical and mechanical stimulation. Physical interactions occur between cancer cells and the surrounding matrix including cell movements and cell shape alterations through the process of mechanotransduction. The latter describes the translation of external mechanical cues into intracellular biochemical signaling. Reorganization of both the cytoskeleton and the extracellular matrix (ECM) plays a critical role in these spreading steps. Migrating tumor cells show increased motility in order to cross the tumor microenvironment, migrate through ECM and reach the bloodstream to the metastatic site. There are specific factors affecting these processes, as well as the survival of circulating tumor cells (CTC) in the blood flow until they finally invade the secondary tissue to form metastasis. This review aims to study the mechanisms of metastasis from a biomechanical perspective and investigate cell migration, with a focus on the alterations in the cytoskeleton through this journey and the effect of biologic fluids on metastasis. Understanding of the biophysical mechanisms that promote tumor metastasis may contribute successful therapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Dimitra Vasilaki
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Alexandros Tsouknidas
- Laboratory for Biomaterials and Computational Mechanics, Department of Mechanical Engineering, University of Western Macedonia, Kozani, Greece
| | | | - Konstantinos Michalakis
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
- Division of Graduate Prosthodontics, Tufts University School of Dental Medicine, Boston, MA USA
- University of Oxford, Oxford, UK
| |
Collapse
|
154
|
Wagh K, Ishikawa M, Garcia DA, Stavreva DA, Upadhyaya A, Hager GL. Mechanical Regulation of Transcription: Recent Advances. Trends Cell Biol 2021; 31:457-472. [PMID: 33712293 PMCID: PMC8221528 DOI: 10.1016/j.tcb.2021.02.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 01/01/2023]
Abstract
Mechanotransduction is the ability of a cell to sense mechanical cues from its microenvironment and convert them into biochemical signals to elicit adaptive transcriptional and other cellular responses. Here, we describe recent advances in the field of mechanical regulation of transcription, highlight mechanical regulation of the epigenome as a key novel aspect of mechanotransduction, and describe recent technological advances that could further elucidate the link between mechanical stimuli and gene expression. In this review, we emphasize the importance of mechanotransduction as one of the governing principles of cancer progression, underscoring the need to conduct further studies of the molecular mechanisms involved in sensing mechanical cues and coordinating transcriptional responses.
Collapse
Affiliation(s)
- Kaustubh Wagh
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Physics, University of Maryland, College Park, MD 20742, USA
| | - Momoko Ishikawa
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David A Garcia
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Physics, University of Maryland, College Park, MD 20742, USA
| | - Diana A Stavreva
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Arpita Upadhyaya
- Department of Physics, University of Maryland, College Park, MD 20742, USA; Institute for Physical Science and Technology, University of Maryland, College Park, MD 20742, USA.
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
155
|
Chirivì M, Maiullari F, Milan M, Presutti D, Cordiglieri C, Crosti M, Sarnicola ML, Soluri A, Volpi M, Święszkowski W, Prati D, Rizzi M, Costantini M, Seliktar D, Parisi C, Bearzi C, Rizzi R. Tumor Extracellular Matrix Stiffness Promptly Modulates the Phenotype and Gene Expression of Infiltrating T Lymphocytes. Int J Mol Sci 2021; 22:5862. [PMID: 34070750 PMCID: PMC8198248 DOI: 10.3390/ijms22115862] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
The immune system is a fine modulator of the tumor biology supporting or inhibiting its progression, growth, invasion and conveys the pharmacological treatment effect. Tumors, on their side, have developed escaping mechanisms from the immune system action ranging from the direct secretion of biochemical signals to an indirect reaction, in which the cellular actors of the tumor microenvironment (TME) collaborate to mechanically condition the extracellular matrix (ECM) making it inhospitable to immune cells. TME is composed of several cell lines besides cancer cells, including tumor-associated macrophages, cancer-associated fibroblasts, CD4+ and CD8+ lymphocytes, and innate immunity cells. These populations interface with each other to prepare a conservative response, capable of evading the defense mechanisms implemented by the host's immune system. The presence or absence, in particular, of cytotoxic CD8+ cells in the vicinity of the main tumor mass, is able to predict, respectively, the success or failure of drug therapy. Among various mechanisms of immunescaping, in this study, we characterized the modulation of the phenotypic profile of CD4+ and CD8+ cells in resting and activated states, in response to the mechanical pressure exerted by a three-dimensional in vitro system, able to recapitulate the rheological and stiffness properties of the tumor ECM.
Collapse
Affiliation(s)
- Maila Chirivì
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, C.so della Repubblica 79, 04100 Latina, Italy
| | - Fabio Maiullari
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Marika Milan
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Via Ercole Ramarini, 32, Monterotondo, 00015 Rome, Italy; (A.S.); (C.P.)
| | - Dario Presutti
- Institute of Physical Chemistry Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224 Warszawa, Poland; (D.P.); (M.C.)
| | - Chiara Cordiglieri
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
| | - Mariacristina Crosti
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
| | - Maria Lucia Sarnicola
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
| | - Andrea Soluri
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Via Ercole Ramarini, 32, Monterotondo, 00015 Rome, Italy; (A.S.); (C.P.)
- Unit of Molecular Neurosciences, University Campus Bio-Medico, 00128 Roma, Italy
| | - Marina Volpi
- Faculty of Materials Science and Engineering, Warsaw University of Technology, 02-507 Warsaw, Poland; (M.V.); (W.Ś.)
| | - Wojciech Święszkowski
- Faculty of Materials Science and Engineering, Warsaw University of Technology, 02-507 Warsaw, Poland; (M.V.); (W.Ś.)
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, IRCCS Granda Hospital Maggiore Policlinico Foundation, Via Francesco Sforza 35, 20122 Milan, Italy;
| | - Marta Rizzi
- Ufficio Programmazione e Grant Office, National Research Council of Italy (UPGO-CNR), Piazzale Aldo Moro 7, 00185 Rome, Italy;
| | - Marco Costantini
- Institute of Physical Chemistry Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224 Warszawa, Poland; (D.P.); (M.C.)
| | - Dror Seliktar
- Department of Biomedical Engineering, Technion Institute, Haifa 32000, Israel;
| | - Chiara Parisi
- Institute of Biochemistry and Cell Biology, National Research Council of Italy (IBBC-CNR), Via Ercole Ramarini, 32, Monterotondo, 00015 Rome, Italy; (A.S.); (C.P.)
| | - Claudia Bearzi
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Institute of Genetic and Biomedical Research, UOS of Milan, National Research Council (IRGB-CNR), Via Gaudenzio Fantoli 16/15, 20138 Milan, Italy
| | - Roberto Rizzi
- Fondazione Istituto Nazionale di Genetica Molecolare, 20122 Milan, Italy; (M.C.); (F.M.); (M.M.); (C.C.); (M.C.); (M.L.S.); (C.B.)
- Institute of Biomedical Technologies, National Research Council (ITB-CNR), Via Fratelli Cervi, 93, Segrate, 20090 Milan, Italy
| |
Collapse
|
156
|
Khalaf K, Hana D, Chou JTT, Singh C, Mackiewicz A, Kaczmarek M. Aspects of the Tumor Microenvironment Involved in Immune Resistance and Drug Resistance. Front Immunol 2021; 12:656364. [PMID: 34122412 PMCID: PMC8190405 DOI: 10.3389/fimmu.2021.656364] [Citation(s) in RCA: 219] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/27/2021] [Indexed: 12/11/2022] Open
Abstract
The tumor microenvironment (TME) is a complex and ever-changing "rogue organ" composed of its own blood supply, lymphatic and nervous systems, stroma, immune cells and extracellular matrix (ECM). These complex components, utilizing both benign and malignant cells, nurture the harsh, immunosuppressive and nutrient-deficient environment necessary for tumor cell growth, proliferation and phenotypic flexibility and variation. An important aspect of the TME is cellular crosstalk and cell-to-ECM communication. This interaction induces the release of soluble factors responsible for immune evasion and ECM remodeling, which further contribute to therapy resistance. Other aspects are the presence of exosomes contributed by both malignant and benign cells, circulating deregulated microRNAs and TME-specific metabolic patterns which further potentiate the progression and/or resistance to therapy. In addition to biochemical signaling, specific TME characteristics such as the hypoxic environment, metabolic derangements, and abnormal mechanical forces have been implicated in the development of treatment resistance. In this review, we will provide an overview of tumor microenvironmental composition, structure, and features that influence immune suppression and contribute to treatment resistance.
Collapse
Affiliation(s)
- Khalil Khalaf
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Doris Hana
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Jadzia Tin-Tsen Chou
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Chandpreet Singh
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Andrzej Mackiewicz
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
157
|
Schoenwaelder N, Salewski I, Engel N, Krause M, Schneider B, Müller M, Riess C, Lemcke H, Skorska A, Grosse-Thie C, Junghanss C, Maletzki C. The Individual Effects of Cyclin-Dependent Kinase Inhibitors on Head and Neck Cancer Cells-A Systematic Analysis. Cancers (Basel) 2021; 13:cancers13102396. [PMID: 34063457 PMCID: PMC8157193 DOI: 10.3390/cancers13102396] [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: 03/01/2021] [Revised: 04/07/2021] [Accepted: 05/11/2021] [Indexed: 12/24/2022] Open
Abstract
Cyclin-dependent kinase inhibitors (CDKi´s) display cytotoxic activity against different malignancies, including head and neck squamous cell carcinomas (HNSCC). By coordinating the DNA damage response, these substances may be combined with cytostatics to enhance cytotoxicity. Here, we investigated the influence of different CDKi´s (palbociclib, dinaciclib, THZ1) on two HNSCC cell lines in monotherapy and combination therapy with clinically-approved drugs (5-FU, Cisplatin, cetuximab). Apoptosis/necrosis, cell cycle, invasiveness, senescence, radiation-induced γ-H2AX DNA double-strand breaks, and effects on the actin filament were studied. Furthermore, the potential to increase tumor immunogenicity was assessed by analyzing Calreticulin translocation and immune relevant surface markers. Finally, an in vivo mouse model was used to analyze the effect of dinaciclib and Cisplatin combination therapy. Dinaciclib, palbociclib, and THZ1 displayed anti-neoplastic activity after low-dose treatment, while the two latter substances slightly enhanced radiosensitivity. Dinaciclib decelerated wound healing, decreased invasiveness, and induced MHC-I, accompanied by high amounts of surface-bound Calreticulin. Numbers of early and late apoptotic cells increased initially (24 h), while necrosis dominated afterward. Antitumoral effects of the selective CDKi palbociclib were weaker, but combinations with 5-FU potentiated effects of the monotherapy. Additionally, CDKi and CDKi/chemotherapy combinations induced MHC I, indicative of enhanced immunogenicity. The in vivo studies revealed a cell line-specific response with best tumor growth control in the combination approach. Global acting CDKi's should be further investigated as targeting agents for HNSCC, either individually or in combination with selected drugs. The ability of dinaciclib to increase the immunogenicity of tumor cells renders this substance a particularly interesting candidate for immune-based oncological treatment regimens.
Collapse
Affiliation(s)
- Nina Schoenwaelder
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
- Correspondence: ; Tel.: +49-381-494-5764
| | - Inken Salewski
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| | - Nadja Engel
- Department of Oral and Maxillofacial Surgery, Facial Plastic Surgery, University Medical Center Rostock, 18057 Rostock, Germany;
| | - Mareike Krause
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| | - Björn Schneider
- Institute of Pathology, University Medical Center Rostock, Strempelstr.14, 18057 Rostock, Germany;
| | - Michael Müller
- Core Facility for Cell Sorting & Cell Analysis, Laboratory for Clinical Immunology, University Medical Center Rostock, 18057 Rostock, Germany;
| | - Christin Riess
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
- University Children’s Hospital, Rostock University Medical Centre, 18057 Rostock, Germany
| | - Heiko Lemcke
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), University Medical Center Rostock, 18057 Rostock, Germany; (H.L.); (A.S.)
- Department of Cardiology, University Medical Center Rostock, 18059 Rostock, Germany
- Department Life, Light & Matter, Faculty of Interdisciplinary Research, University Rostock, 18059 Rostock, Germany
| | - Anna Skorska
- Department of Cardiac Surgery, Reference and Translation Center for Cardiac Stem Cell Therapy (RTC), University Medical Center Rostock, 18057 Rostock, Germany; (H.L.); (A.S.)
- Department of Cardiology, University Medical Center Rostock, 18059 Rostock, Germany
- Department Life, Light & Matter, Faculty of Interdisciplinary Research, University Rostock, 18059 Rostock, Germany
| | - Christina Grosse-Thie
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| | - Christian Junghanss
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| | - Claudia Maletzki
- Department of Internal Medicine, Medical Clinic III—Hematology, Oncology, Palliative Medicine, University Medical Center Rostock, 18057 Rostock, Germany; (I.S.); (M.K.); (C.R.); (C.G.-T.); (C.J.); (C.M.)
| |
Collapse
|
158
|
Xuan B, Ghosh D, Dawson MR. Contributions of the distinct biophysical phenotype of polyploidal giant cancer cells to cancer progression. Semin Cancer Biol 2021; 81:64-72. [PMID: 33992783 DOI: 10.1016/j.semcancer.2021.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 01/27/2023]
Abstract
Polyploid giant cancer cells (PGCCs) are a commonly observed histological feature of human tumors and are particularly prominent in late stage and drug resistant cancers. The chromosomal duplication conferred by their aneuploidy gives rise to DNA damage resistance and complex tumor cell karyotypes, a driving factor in chemotherapy resistance and disease relapse. Furthermore, PGCCs also exhibit key cytoskeletal features that give rise to a distinct biophysical phenotype, including increased density of polymerized actin and vimentin intermediate filaments, nuclear and cytoskeletal stiffening, increased traction force, and migratory persistence. Despite recent research highlighting the role PGCCs play in cancer progression, this population of tumor cells remains poorly characterized in terms of their biophysical properties. In this review, we will discuss the various aspects of their biomolecular phenotype, such as increased stemness as well as a mixed EMT signature. These features have been extensively associated with tumorigenesis and recurrence, and aggressive cancers. Additionally, we will also examine the distinct PGCC cytoskeletal features of actin and filamentous vimentin. Specifically, how the differential organization of these networks serve to support their increased size and drive migratory persistence. These findings could shed light on potential therapeutic strategies that allow for specific elimination or mitigation of the invasive potential of these polyploid cancer cells. Lastly, we will examine how the biophysical and molecular phenotype of PGCCs combine to tip the scale in favor of promoting cancer progression, presenting an important target in the clinical treatment of cancer.
Collapse
Affiliation(s)
- Botai Xuan
- Brown University, Department of Molecular Pharmacology, Physiology, & Biotechnology, Providence, 02912, USA
| | - Deepraj Ghosh
- Brown University, Department of Molecular Pharmacology, Physiology, & Biotechnology, Providence, 02912, USA
| | - Michelle R Dawson
- Brown University, Department of Molecular Pharmacology, Physiology, & Biotechnology, Providence, 02912, USA; Brown University, Center for Biomedical Engineering, Providence, 02912, USA; Brown University, School of Engineering, Providence, 02912, USA.
| |
Collapse
|
159
|
Fuentes-Chandía M, Vierling A, Kappelmann-Fenzl M, Monavari M, Letort G, Höne L, Parma B, Antara SK, Ertekin Ö, Palmisano R, Dong M, Böpple K, Boccaccini AR, Ceppi P, Bosserhoff AK, Leal-Egaña A. 3D Spheroids Versus 3D Tumor-Like Microcapsules: Confinement and Mechanical Stress May Lead to the Expression of Malignant Responses in Cancer Cells. Adv Biol (Weinh) 2021; 5:e2000349. [PMID: 33960743 DOI: 10.1002/adbi.202000349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 04/03/2021] [Indexed: 11/08/2022]
Abstract
As 2D surfaces fail to resemble the tumoral milieu, current discussions are focused on which 3D cell culture strategy may better lead the cells to express in vitro most of the malignant hints described in vivo. In this study, this question is assessed by analyzing the full genetic profile of MCF7 cells cultured either as 3D spheroids-considered as "gold standard" for in vitro cancer research- or immobilized in 3D tumor-like microcapsules, by RNA-Seq and transcriptomic methods, allowing to discriminate at big-data scale, which in vitro strategy can better resemble most of the malignant features described in neoplastic diseases. The results clearly show that mechanical stress, rather than 3D morphology only, stimulates most of the biological processes involved in cancer pathogenicity, such as cytoskeletal organization, migration, and stemness. Furthermore, cells entrapped in hydrogel-based scaffolds are likely expressing other physiological hints described in malignancy, such as the upregulated expression of metalloproteinases or the resistance to anticancer drugs, among others. According to the knowledge, this study represents the first attempt to answer which 3D experimental system can better mimic the neoplastic architecture in vitro, emphasizing the relevance of confinement in cancer pathogenicity, which can be easily achieved by using hydrogel-based matrices.
Collapse
Affiliation(s)
- Miguel Fuentes-Chandía
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Andreas Vierling
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Melanie Kappelmann-Fenzl
- Institute of Biochemistry, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, Fahrstraße 17, 91054, Erlangen, Germany.,Faculty of Applied Informatics, University of Applied Science Deggendorf, 94469, Deggendorf, Germany
| | - Mahshid Monavari
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Gaelle Letort
- Center for Interdisciplinary Research in Biology, Collège de France UMR7241/U1050. 11, place Marcelin Berthelot, Paris Cedex 05, 75231, France
| | - Lucas Höne
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Beatrice Parma
- Interdisciplinary Center for Clinical Research (IZKF), Friedrich-Alexander University of Erlangen-Nuremberg, Glueckstraße 6, 91054, Erlangen, Germany
| | - Sharmin Khan Antara
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Özlem Ertekin
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Ralph Palmisano
- Optical Imaging Centre Erlangen, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 3, 91058, Erlangen, Germany
| | - Meng Dong
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and University of Tübingen, Auerbachstraße 112, 70376, Stuttgart, Germany
| | - Kathrin Böpple
- Dr. Margarete Fischer-Bosch-Institute of Clinical Pharmacology and University of Tübingen, Auerbachstraße 112, 70376, Stuttgart, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, 91058, Erlangen, Germany
| | - Paolo Ceppi
- Interdisciplinary Center for Clinical Research (IZKF), Friedrich-Alexander University of Erlangen-Nuremberg, Glueckstraße 6, 91054, Erlangen, Germany.,Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense M, DK-5230, Denmark
| | - Anja K Bosserhoff
- Institute of Biochemistry, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Aldo Leal-Egaña
- Institute of Biomaterials, Friedrich-Alexander Universität Erlangen-Nürnberg, Cauerstraße 6, 91058, Erlangen, Germany.,Institute for Molecular Systems Engineering, Heidelberg University, In Neuenheimer Feld 253, 69120, Heidelberg, Germany
| |
Collapse
|
160
|
Wu Y, Zanotelli MR, Zhang J, Reinhart-King CA. Matrix-driven changes in metabolism support cytoskeletal activity to promote cell migration. Biophys J 2021; 120:1705-1717. [PMID: 33705759 PMCID: PMC8204337 DOI: 10.1016/j.bpj.2021.02.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 02/03/2021] [Accepted: 02/23/2021] [Indexed: 01/21/2023] Open
Abstract
The microenvironment provides both active and passive mechanical cues that regulate cell morphology, adhesion, migration, and metabolism. Although the cellular response to those mechanical cues often requires energy-intensive actin cytoskeletal remodeling and actomyosin contractility, it remains unclear how cells dynamically adapt their metabolic activity to altered mechanical cues to support migration. Here, we investigated the changes in cellular metabolic activity in response to different two-dimensional and three-dimensional microenvironmental conditions and how these changes relate to cytoskeletal activity and migration. Utilizing collagen micropatterning on polyacrylamide gels, intracellular energy levels and oxidative phosphorylation were found to be correlated with cell elongation and spreading and necessary for membrane ruffling. To determine whether this relationship holds in more physiological three-dimensional matrices, collagen matrices were used to show that intracellular energy state was also correlated with protrusive activity and increased with matrix density. Pharmacological inhibition of oxidative phosphorylation revealed that cancer cells rely on oxidative phosphorylation to meet the elevated energy requirements for protrusive activity and migration in denser matrices. Together, these findings suggest that mechanical regulation of cytoskeletal activity during spreading and migration by the physical microenvironment is driven by an altered metabolic profile.
Collapse
Affiliation(s)
- Yusheng Wu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Matthew R Zanotelli
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jian Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | |
Collapse
|
161
|
Sherekar S, Viswanathan GA. Boolean dynamic modeling of cancer signaling networks: Prognosis, progression, and therapeutics. COMPUTATIONAL AND SYSTEMS ONCOLOGY 2021. [DOI: 10.1002/cso2.1017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Shubhank Sherekar
- Department of Chemical Engineering Indian Institute of Technology Bombay, Powai Mumbai India
| | - Ganesh A. Viswanathan
- Department of Chemical Engineering Indian Institute of Technology Bombay, Powai Mumbai India
| |
Collapse
|
162
|
Pandkar MR, Dhamdhere SG, Shukla S. Oxygen gradient and tumor heterogeneity: The chronicle of a toxic relationship. Biochim Biophys Acta Rev Cancer 2021; 1876:188553. [PMID: 33915221 DOI: 10.1016/j.bbcan.2021.188553] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/08/2021] [Accepted: 04/21/2021] [Indexed: 12/21/2022]
Abstract
The commencement of cancer is attributed to one or a few cells that become rogue and attain the property of immortality. The inception of distinct cancer cell clones during the hyperplastic and dysplastic stages of cancer progression is the utimate consequence of the dysregulated cellular pathways and the proliferative potential itself. Furthermore, a critical factor that adds a layer of complexity to this pre-existent intra-tumoral heterogeneity (ITH) is the foundation of an oxygen gradient, that is established due to the improper architecture of the tumor vasculature. Therefore, as a resultant effect, the poorly oxygenated regions thus formed and characterized as hypoxic, promote the emergence of aggressive and treatment-resistant cancer cell clones. The extraordinary property of the hypoxic cancer cells to exist harmoniously with cancerous and non-cancerous cells in the tumor microenvironment (TME) further increases the intricacies of ITH. Here in this review, the pivotal influence of differential oxygen concentrations in shaping the ITH is thoroughly discussed. We also emphasize on the vitality of the interacting networks that govern the overall fate of oxygen gradient-dependent origin of tumor heterogeneity. Additionally, the implications of less-appreciated reverse Warburg effect, a symbiotic metabolic coupling, and the associated epigenetic regulation of rewiring of cancer metabolism in response to oxygen gradients, have been highlighted as critical influencers of ITH.
Collapse
Affiliation(s)
- Madhura R Pandkar
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh 462066, India
| | - Shruti G Dhamdhere
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh 462066, India
| | - Sanjeev Shukla
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, Madhya Pradesh 462066, India.
| |
Collapse
|
163
|
Fabrication of Adhesive Substrate for Incorporating Hydrogels to Investigate the Influence of Stiffness on Cancer Cell Behavior. Methods Mol Biol 2021; 2174:277-297. [PMID: 32813257 DOI: 10.1007/978-1-0716-0759-6_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Stiffness control of cell culture platforms provides researchers in cell biology with the ability to study different experimental models in conditions of mimicking physiological or pathological microenvironments. Nevertheless, the signal transduction pathways and drug sensibility of cancer cells have been poorly characterized widely using biomimetic platforms because the limited experience of cancer cell biology groups about handling substrates with specific mechanical properties. The protein cross-linking and stiffening control are crucial checkpoints that could strongly affect cell adhesion and spreading, misrepresenting the data acquired, and also generating inaccurate cellular models. Here, we introduce a simple method to adhere to polyacrylamide (PAA) hydrogels on glass coverslips without any special treatment for mechanics studies in cancer cell biology. By using a commercial photosensitive glue, Loctite 3525, it is possible to polymerize PAA hydrogels directly on glass surfaces. Furthermore, we describe a cross-linking reaction method to attach proteins to PAA as an alternative method to Sulfo-SANPAH cross-linking, which is sometimes difficult to implement and reproduce. In this chapter, we describe a reliable procedure to fabricate ECM protein-cross-linked PAA hydrogels for mechanotransduction studies on cancer cells.
Collapse
|
164
|
Chen Y, Kim J, Yang S, Wang H, Wu CJ, Sugimoto H, LeBleu VS, Kalluri R. Type I collagen deletion in αSMA + myofibroblasts augments immune suppression and accelerates progression of pancreatic cancer. Cancer Cell 2021; 39:548-565.e6. [PMID: 33667385 PMCID: PMC8423173 DOI: 10.1016/j.ccell.2021.02.007] [Citation(s) in RCA: 301] [Impact Index Per Article: 100.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/23/2020] [Accepted: 02/10/2021] [Indexed: 12/11/2022]
Abstract
Stromal desmoplastic reaction in pancreatic ductal adenocarcinoma (PDAC) involves significant accumulation of type I collagen (Col1). However, the precise molecular and mechanistic contribution of Col1 in PDAC progression remains unknown. Activated pancreatic stellate cells/αSMA+ myofibroblasts are major contributors of Col1 in the PDAC stroma. We use a dual-recombinase genetic mouse model of spontaneous PDAC to delete Col1 specifically in myofibroblasts. This results in significant reduction of total stromal Col1 content and accelerates the emergence of PanINs and PDAC, decreasing overall survival. Col1 deletion leads to Cxcl5 upregulation in cancer cells via SOX9. Increase in Cxcl5 is associated with recruitment of myeloid-derived suppressor cells and suppression of CD8+ T cells, which can be attenuated with combined targeting of CXCR2 and CCR2 to restrain accelerated PDAC progression in the setting of stromal Col1 deletion. Our results unravel the fundamental role of myofibroblast-derived Co1l in regulating tumor immunity and restraining PDAC progression.
Collapse
Affiliation(s)
- Yang Chen
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jiha Kim
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Sujuan Yang
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Huamin Wang
- Department of Anatomical Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Chang-Jiun Wu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Hikaru Sugimoto
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Valerie S LeBleu
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
165
|
Acharya BR. Can mechanical forces attune heterotypic cell-cell communications? J Biomech 2021; 121:110409. [PMID: 33845355 DOI: 10.1016/j.jbiomech.2021.110409] [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: 11/30/2020] [Revised: 02/18/2021] [Accepted: 02/22/2021] [Indexed: 10/21/2022]
Abstract
Heterotypic cell lineages relentlessly exchange biomechanical signals among themselves in metazoan organs. Hence, cell-cell communications are pivotal for organ physiology and pathogenesis. Every cell lineage of an organ responds differently to a specific signal due to its unique receptibility and signal interpretation capacity. These distinct cellular responses generate a system-scale signaling network that helps in generating a specific organ phenotype. Although the reciprocal biochemical signal exchange between non-identical neighboring cells is known to be an essential factor for organ functioning, if, then how, mechanical cues incite these signals is not yet quite explored. Cells within organ tissues experience multiple mechanical forces, such as stretching, bending, compression, and shear stress. Forms and magnitudes of mechanical forces influence biochemical signaling in a cell-specific manner. Additionally, the biophysical state of acellular extracellular matrix (ECM) can transmit exclusive mechanical cues to specific cells of an organ. As it scaffolds heterotypic cells and tissues in close proximities, therefore, ECM can easily be contemplated as a mechanical conduit for signal exchange among them. However, force-stimulated signal transduction is not always physiological, aberrant force sensing by tissue-resident cells can transduce anomalous signals to each other, and potentially can promote pathological phenotypes. Herein, I attempt to put forward a perspective on how mechanical forces may influence signal transductions among heterotypic cell populations and how they feedback each other to achieve a transient or perpetual alteration in metazoan organs. A mechanistic insight of organ scale mechanotransduction can emanate the possibility of finding potential biomarkers and novel therapeutic strategies to deal with pathogenesis and organ regeneration.
Collapse
Affiliation(s)
- Bipul R Acharya
- Department of Cell Biology, School of Medicine, University of Virginia, USA.
| |
Collapse
|
166
|
Location, location, location: how the tissue microenvironment affects inflammation in RA. Nat Rev Rheumatol 2021; 17:195-212. [PMID: 33526927 DOI: 10.1038/s41584-020-00570-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 01/30/2023]
Abstract
Current treatments for rheumatoid arthritis (RA) do not work well for a large proportion of patients, or at all in some individuals, and cannot cure or prevent this disease. One major obstacle to developing better drugs is a lack of complete understanding of how inflammatory joint disease arises and progresses. Emerging evidence indicates an important role for the tissue microenvironment in the pathogenesis of RA. Each tissue is made up of cells surrounded and supported by a unique extracellular matrix (ECM). These complex molecular networks define tissue architecture and provide environmental signals that programme site-specific cell behaviour. In the synovium, a main site of disease activity in RA, positional and disease stage-specific cellular diversity exist. Improved understanding of the architecture of the synovium from gross anatomy to the single-cell level, in parallel with evidence demonstrating how the synovial ECM is vital for synovial homeostasis and how dysregulated signals from the ECM promote chronic inflammation and tissue destruction in the RA joint, has opened up new ways of thinking about the pathogenesis of RA. These new ideas provide novel therapeutic approaches for patients with difficult-to-treat disease and could also be used in disease prevention.
Collapse
|
167
|
Nemec S, Lam J, Zhong J, Heu C, Timpson P, Li Q, Youkhana J, Sharbeen G, Phillips PA, Kilian KA. Interfacial Curvature in Confined Coculture Directs Stromal Cell Activity with Spatial Corralling of Pancreatic Cancer Cells. Adv Biol (Weinh) 2021; 5:e2000525. [PMID: 33754491 DOI: 10.1002/adbi.202000525] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/18/2021] [Indexed: 01/18/2023]
Abstract
Interfacial cues in the tumor microenvironment direct the activity and assembly of multiple cell types. Pancreatic cancer, along with breast and prostate cancers, is enriched with cancer-associated fibroblasts (CAFs) that activate to coordinate the deposition of the extracellular matrix, which can comprise over 90% of the tumor mass. While it is clear that matrix underlies the severity of the disease, the relationship between stromal-tumor cell assembly and cell-matrix dynamics remains elusive. Micropatterned hydrogels deconstruct the interplay between matrix stiffness and geometric confinement, guiding heterotypic cell populations and matrix assembly in pancreatic cancer. Interfacial cues at the perimeter of microislands guide CAF migration and direct cancer cell assembly. Computational modeling shows curvature-stress dependent cellular localization for cancer and CAFs in coculture. Regions of convex curvature enhance edge stress that activates a myofibroblast phenotype in the CAFs with migration and increased collagen I deposition, ultimately leading to a central "corralling" of cancer cells. Inhibiting mechanotransduction pathways decreases CAF activation and the associated corralling phenotype. Together, this work reveals how interfacial biophysical cues underpin aspects of stromal desmoplasia, a hallmark of disease severity and chemoresistance in the pancreatic, breast, and prostate cancers, thereby providing a tool to expand stroma-targeting therapeutic strategies.
Collapse
Affiliation(s)
- Stephanie Nemec
- School of Materials Science and Engineering, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Joey Lam
- School of Chemistry Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Jingxiao Zhong
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Celine Heu
- Biomedical Imaging Facility, Mark Wainwright Analytical Center, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Paul Timpson
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, 2052, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Janet Youkhana
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Phoebe A Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Sciences, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Kristopher A Kilian
- School of Materials Science and Engineering, UNSW Sydney, Sydney, NSW, 2052, Australia.,School of Chemistry Australian Centre for Nanomedicine, UNSW Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|
168
|
Tello-Lafoz M, Srpan K, Sanchez EE, Hu J, Remsik J, Romin Y, Calò A, Hoen D, Bhanot U, Morris L, Boire A, Hsu KC, Massagué J, Huse M, Er EE. Cytotoxic lymphocytes target characteristic biophysical vulnerabilities in cancer. Immunity 2021; 54:1037-1054.e7. [PMID: 33756102 DOI: 10.1016/j.immuni.2021.02.020] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 01/16/2021] [Accepted: 02/25/2021] [Indexed: 12/17/2022]
Abstract
Immune cells identify and destroy tumors by recognizing cellular traits indicative of oncogenic transformation. In this study, we found that myocardin-related transcription factors (MRTFs), which promote migration and metastatic invasion, also sensitize cancer cells to the immune system. Melanoma and breast cancer cells with high MRTF expression were selectively eliminated by cytotoxic lymphocytes in mouse models of metastasis. This immunosurveillance phenotype was further enhanced by treatment with immune checkpoint blockade (ICB) antibodies. We also observed that high MRTF signaling in human melanoma is associated with ICB efficacy in patients. Using biophysical and functional assays, we showed that MRTF overexpression rigidified the filamentous actin cytoskeleton and that this mechanical change rendered mouse and human cancer cells more vulnerable to cytotoxic T lymphocytes and natural killer cells. Collectively, these results suggest that immunosurveillance has a mechanical dimension, which we call mechanosurveillance, that is particularly relevant for the targeting of metastatic disease.
Collapse
Affiliation(s)
- Maria Tello-Lafoz
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katja Srpan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa E Sanchez
- Biochemistry and Molecular Biology Program, Weill Cornell Medical College, New York, NY, USA
| | - Jing Hu
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jan Remsik
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yevgeniy Romin
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Annalisa Calò
- Molecular Cytology Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Douglas Hoen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Umeshkumar Bhanot
- Precision Pathology Center, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Luc Morris
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Adrienne Boire
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katharine C Hsu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joan Massagué
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Morgan Huse
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Ekrem Emrah Er
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
169
|
Architectural control of metabolic plasticity in epithelial cancer cells. Commun Biol 2021; 4:371. [PMID: 33742081 PMCID: PMC7979883 DOI: 10.1038/s42003-021-01899-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 02/15/2021] [Indexed: 12/11/2022] Open
Abstract
Metabolic plasticity enables cancer cells to switch between glycolysis and oxidative phosphorylation to adapt to changing conditions during cancer progression, whereas metabolic dependencies limit plasticity. To understand a role for the architectural environment in these processes we examined metabolic dependencies of cancer cells cultured in flat (2D) and organotypic (3D) environments. Here we show that cancer cells in flat cultures exist in a high energy state (oxidative phosphorylation), are glycolytic, and depend on glucose and glutamine for growth. In contrast, cells in organotypic culture exhibit lower energy and glycolysis, with extensive metabolic plasticity to maintain growth during glucose or amino acid deprivation. Expression of KRASG12V in organotypic cells drives glucose dependence, however cells retain metabolic plasticity to glutamine deprivation. Finally, our data reveal that mechanical properties control metabolic plasticity, which correlates with canonical Wnt signaling. In summary, our work highlights that the architectural and mechanical properties influence cells to permit or restrict metabolic plasticity.
Collapse
|
170
|
Benítez L, Barberis L, Vellón L, Condat CA. Understanding the influence of substrate when growing tumorspheres. BMC Cancer 2021; 21:276. [PMID: 33722191 PMCID: PMC7962376 DOI: 10.1186/s12885-021-07918-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 02/15/2021] [Indexed: 11/18/2022] Open
Abstract
Background Cancer stem cells are important for the development of many solid tumors. These cells receive promoting and inhibitory signals that depend on the nature of their environment (their niche) and determine cell dynamics. Mechanical stresses are crucial to the initiation and interpretation of these signals. Methods A two-population mathematical model of tumorsphere growth is used to interpret the results of a series of experiments recently carried out in Tianjin, China, and extract information about the intraspecific and interspecific interactions between cancer stem cell and differentiated cancer cell populations. Results The model allows us to reconstruct the time evolution of the cancer stem cell fraction, which was not directly measured. We find that, in the presence of stem cell growth factors, the interspecific cooperation between cancer stem cells and differentiated cancer cells induces a positive feedback loop that determines growth, independently of substrate hardness. In a frustrated attempt to reconstitute the stem cell niche, the number of cancer stem cells increases continuously with a reproduction rate that is enhanced by a hard substrate. For growth on soft agar, intraspecific interactions are always inhibitory, but on hard agar the interactions between stem cells are collaborative while those between differentiated cells are strongly inhibitory. Evidence also suggests that a hard substrate brings about a large fraction of asymmetric stem cell divisions. In the absence of stem cell growth factors, the barrier to differentiation is broken and overall growth is faster, even if the stem cell number is conserved. Conclusions Our interpretation of the experimental results validates the centrality of the concept of stem cell niche when tumor growth is fueled by cancer stem cells. Niche memory is found to be responsible for the characteristic population dynamics observed in tumorspheres. The model also shows why substratum stiffness has a deep influence on the behavior of cancer stem cells, stiffer substrates leading to a larger proportion of asymmetric doublings. A specific condition for the growth of the cancer stem cell number is also obtained Supplementary Information The online version contains supplementary material available at (10.1186/s12885-021-07918-1).
Collapse
Affiliation(s)
- Lucía Benítez
- Instituto de Física Enrique Gaviola, CONICET, and Facultad de Matemática, Astronomía, Física y Computación, Universidad Nacional de Córdoba, Córdoba, X5000 HUA, Argentina
| | - Lucas Barberis
- Instituto de Física Enrique Gaviola, CONICET, and Facultad de Matemática, Astronomía, Física y Computación, Universidad Nacional de Córdoba, Córdoba, X5000 HUA, Argentina.
| | - Luciano Vellón
- Instituto de Biología y Medicina Experimental, CONICET., Buenos Aires, C1428 ADN, Argentina
| | - Carlos A Condat
- Instituto de Física Enrique Gaviola, CONICET, and Facultad de Matemática, Astronomía, Física y Computación, Universidad Nacional de Córdoba, Córdoba, X5000 HUA, Argentina
| |
Collapse
|
171
|
Advanced Spheroid, Tumouroid and 3D Bioprinted In-Vitro Models of Adult and Paediatric Glioblastoma. Int J Mol Sci 2021; 22:ijms22062962. [PMID: 33803967 PMCID: PMC8000246 DOI: 10.3390/ijms22062962] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 12/16/2022] Open
Abstract
The life expectancy of patients with high-grade glioma (HGG) has not improved in decades. One of the crucial tools to enable future improvement is advanced models that faithfully recapitulate the tumour microenvironment; they can be used for high-throughput screening that in future may enable accurate personalised drug screens. Currently, advanced models are crucial for identifying and understanding potential new targets, assessing new chemotherapeutic compounds or other treatment modalities. Recently, various methodologies have come into use that have allowed the validation of complex models—namely, spheroids, tumouroids, hydrogel-embedded cultures (matrix-supported) and advanced bioengineered cultures assembled with bioprinting and microfluidics. This review is designed to present the state of advanced models of HGG, whilst focusing as much as is possible on the paediatric form of the disease. The reality remains, however, that paediatric HGG (pHGG) models are years behind those of adult HGG. Our goal is to bring this to light in the hope that pGBM models can be improved upon.
Collapse
|
172
|
Graybill PM, Bollineni RK, Sheng Z, Davalos RV, Mirzaeifar R. A constriction channel analysis of astrocytoma stiffness and disease progression. BIOMICROFLUIDICS 2021; 15:024103. [PMID: 33763160 PMCID: PMC7968935 DOI: 10.1063/5.0040283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/23/2021] [Indexed: 05/12/2023]
Abstract
Studies have demonstrated that cancer cells tend to have reduced stiffness (Young's modulus) compared to their healthy counterparts. The mechanical properties of primary brain cancer cells, however, have remained largely unstudied. To investigate whether the stiffness of primary brain cancer cells decreases as malignancy increases, we used a microfluidic constriction channel device to deform healthy astrocytes and astrocytoma cells of grade II, III, and IV and measured the entry time, transit time, and elongation. Calculating cell stiffness directly from the experimental measurements is not possible. To overcome this challenge, finite element simulations of the cell entry into the constriction channel were used to train a neural network to calculate the stiffness of the analyzed cells based on their experimentally measured diameter, entry time, and elongation in the channel. Our study provides the first calculation of stiffness for grades II and III astrocytoma and is the first to apply a neural network analysis to determine cell mechanical properties from a constriction channel device. Our results suggest that the stiffness of astrocytoma cells is not well-correlated with the cell grade. Furthermore, while other non-central-nervous-system cell types typically show reduced stiffness of malignant cells, we found that most astrocytoma cell lines had increased stiffness compared to healthy astrocytes, with lower-grade astrocytoma having higher stiffness values than grade IV glioblastoma. Differences in nucleus-to-cytoplasm ratio only partly explain differences in stiffness values. Although our study does have limitations, our results do not show a strong correlation of stiffness with cell grade, suggesting that other factors may play important roles in determining the invasive capability of astrocytoma. Future studies are warranted to further elucidate the mechanical properties of astrocytoma across various pathological grades.
Collapse
Affiliation(s)
| | - R. K. Bollineni
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Z. Sheng
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine and Virginia Tech Fralin Biomedical Research Institute, Roanoke, Virginia 24016, USA
| | - R. V. Davalos
- Authors to whom correspondence should be addressed: and
| | - R. Mirzaeifar
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, Virginia 24061, USA
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
173
|
Hernández-Cáceres MP, Munoz L, Pradenas JM, Pena F, Lagos P, Aceiton P, Owen GI, Morselli E, Criollo A, Ravasio A, Bertocchi C. Mechanobiology of Autophagy: The Unexplored Side of Cancer. Front Oncol 2021; 11:632956. [PMID: 33718218 PMCID: PMC7952994 DOI: 10.3389/fonc.2021.632956] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Proper execution of cellular function, maintenance of cellular homeostasis and cell survival depend on functional integration of cellular processes and correct orchestration of cellular responses to stresses. Cancer transformation is a common negative consequence of mismanagement of coordinated response by the cell. In this scenario, by maintaining the balance among synthesis, degradation, and recycling of cytosolic components including proteins, lipids, and organelles the process of autophagy plays a central role. Several environmental stresses activate autophagy, among those hypoxia, DNA damage, inflammation, and metabolic challenges such as starvation. In addition to these chemical challenges, there is a requirement for cells to cope with mechanical stresses stemming from their microenvironment. Cells accomplish this task by activating an intrinsic mechanical response mediated by cytoskeleton active processes and through mechanosensitive protein complexes which interface the cells with their mechano-environment. Despite autophagy and cell mechanics being known to play crucial transforming roles during oncogenesis and malignant progression their interplay is largely overlooked. In this review, we highlight the role of physical forces in autophagy regulation and their potential implications in both physiological as well as pathological conditions. By taking a mechanical perspective, we wish to stimulate novel questions to further the investigation of the mechanical requirements of autophagy and appreciate the extent to which mechanical signals affect this process.
Collapse
Affiliation(s)
- Maria Paz Hernández-Cáceres
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Leslie Munoz
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Javiera M. Pradenas
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Pena
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Lagos
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Pablo Aceiton
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| | - Gareth I. Owen
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Investigation in Oncology, Faculty of Biological Sciences Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Eugenia Morselli
- Laboratory of Autophagy and Metabolism, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
| | - Alfredo Criollo
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Autophagy Research Center, Santiago de Chile, Chile
- Facultad De Odontología, Instituto De Investigación En Ciencias Odontológicas (ICOD), Universidad De Chile, Santiago, Chile
| | - Andrea Ravasio
- Laboratory for Mechanobiology of Transforming Systems, Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristina Bertocchi
- Laboratory for Molecular Mechanics of Cell Adhesion, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica De Chile, Santiago, Chile
| |
Collapse
|
174
|
Piezo 1 activation facilitates cholangiocarcinoma metastasis via Hippo/YAP signaling axis. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 24:241-252. [PMID: 33767919 PMCID: PMC7973248 DOI: 10.1016/j.omtn.2021.02.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
Abstract
Tumor metastasis is one of the major factors for the high mortality in cholangiocarcinoma (CCA), but its underlying mechanisms are not fully understood. Here, we report that Piezo-type mechanosensitive ion channel component 1 (Piezo 1) is detected to be significantly upregulated in CCA tissues, which is linked to a poor prognosis in patients, suggesting that Piezo 1 may act in a pro-metastatic role in CCA development. Piezo 1 is activated through 20% simulated physiological stretch, and deleting Piezo 1 impedes epithelial-to-mesenchymal transition (EMT) of CCA cells, as well as impairing their metastatic capacity in vitro and in vivo. Mechanistically, the activation of Piezo 1 results in large amounts of Yes-associated protein 1 (YAP) translocated into the nucleus from the cytoplasm, and thus the motility of CCA cells is significantly increased. These findings indicate that mechanical stimulation induces Piezo 1 activation, which might be involved in CCA metastasis via the Hippo/YAP signaling axis. Therefore, Piezo 1 and its downstream effectors may be a novel therapeutic target for CCA treatment.
Collapse
|
175
|
Stiffness increases with myofibroblast content and collagen density in mesenchymal high grade serous ovarian cancer. Sci Rep 2021; 11:4219. [PMID: 33603134 PMCID: PMC7892556 DOI: 10.1038/s41598-021-83685-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/05/2021] [Indexed: 01/31/2023] Open
Abstract
Women diagnosed with high-grade serous ovarian cancers (HGSOC) are still likely to exhibit a bad prognosis, particularly when suffering from HGSOC of the Mesenchymal molecular subtype (50% cases). These tumors show a desmoplastic reaction with accumulation of extracellular matrix proteins and high content of cancer-associated fibroblasts. Using patient-derived xenograft mouse models of Mesenchymal and Non-Mesenchymal HGSOC, we show here that HGSOC exhibit distinct stiffness depending on their molecular subtype. Indeed, tumor stiffness strongly correlates with tumor growth in Mesenchymal HGSOC, while Non-Mesenchymal tumors remain soft. Moreover, we observe that tumor stiffening is associated with high stromal content, collagen network remodeling, and MAPK/MEK pathway activation. Furthermore, tumor stiffness accompanies a glycolytic metabolic switch in the epithelial compartment, as expected based on Warburg's effect, but also in stromal cells. This effect is restricted to the central part of stiff Mesenchymal tumors. Indeed, stiff Mesenchymal tumors remain softer at the periphery than at the core, with stromal cells secreting high levels of collagens and showing an OXPHOS metabolism. Thus, our study suggests that tumor stiffness could be at the crossroad of three major processes, i.e. matrix remodeling, MEK activation and stromal metabolic switch that might explain at least in part Mesenchymal HGSOC aggressiveness.
Collapse
|
176
|
Redmond J, McCarthy H, Buchanan P, Levingstone TJ, Dunne NJ. Advances in biofabrication techniques for collagen-based 3D in vitro culture models for breast cancer research. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 122:111944. [PMID: 33641930 DOI: 10.1016/j.msec.2021.111944] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/19/2022]
Abstract
Collagen is the most abundant component of the extracellular matrix (ECM), therefore it represents an ideal biomaterial for the culture of a variety of cell types. Recently, collagen-based scaffolds have shown promise as 3D culture platforms for breast cancer-based research. Two-dimensional (2D) in vitro culture models, while useful for gaining preliminary insights, are ultimately flawed as they do not adequately replicate the tumour microenvironment. As a result, they do not facilitate proper 3D cell-cell/cell-matrix interactions and often an exaggerated response to therapeutic agents occurs. The ECM plays a crucial role in the development and spread of cancer. Alterations within the ECM have a significant impact on the pathogenesis of cancer, the initiation of metastasis and ultimate progression of the disease. 3D in vitro culture models that aim to replicate the tumour microenvironment have the potential to offer a new frontier for cancer research with cell growth, morphology and genetic properties that more closely match in vivo cancers. While initial 3D in vitro culture models used in breast cancer research consisted of simple hydrogel platforms, recent advances in biofabrication techniques, including freeze-drying, electrospinning and 3D bioprinting, have enabled the fabrication of biomimetic collagen-based platforms that more closely replicate the breast cancer ECM. This review highlights the current application of collagen-based scaffolds as 3D in vitro culture models for breast cancer research, specifically for adherence-based scaffolds (i.e. matrix-assisted). Finally, the future perspectives of 3D in vitro breast cancer models and their potential to lead to an improved understanding of breast cancer diagnosis and treatment are discussed.
Collapse
Affiliation(s)
- John Redmond
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland
| | - Helen McCarthy
- School of Pharmacy, Queen's University, Belfast BT9 7BL, United Kingdom; School of Chemical Sciences, Dublin City University, Dublin 9, Ireland
| | - Paul Buchanan
- School of Nursing and Human Science, Dublin City University, Dublin 9, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Tanya J Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland; Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Centre for Medical Engineering Research, Dublin City University, Dublin 9, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
177
|
Kulwatno J, Gearhart J, Gong X, Herzog N, Getzin M, Skobe M, Mills KL. Growth of tumor emboli within a vessel model reveals dependence on the magnitude of mechanical constraint. Integr Biol (Camb) 2021; 13:1-16. [PMID: 33443535 DOI: 10.1093/intbio/zyaa024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/02/2020] [Accepted: 12/03/2020] [Indexed: 01/18/2023]
Abstract
Tumor emboli-aggregates of tumor cells within vessels-pose a clinical challenge as they are associated with increased metastasis and tumor recurrence. When growing within a vessel, tumor emboli are subject to a unique mechanical constraint provided by the tubular geometry of the vessel. Current models of tumor emboli use unconstrained multicellular tumor spheroids, which neglect this mechanical interplay. Here, we modeled a lymphatic vessel as a 200 μm-diameter channel in either a stiff or soft, bioinert agarose matrix to create a vessel-like constraint model (VLCM), and we modeled colon or breast cancer tumor emboli with aggregates of HCT116 or SUM149PT cells, respectively. The stiff matrix VLCM constrained the tumor emboli to the cylindrical channel, which led to continuous growth of the emboli, in contrast to the growth rate reduction that unconstrained spheroids exhibit. Emboli morphology in the soft matrix VLCM, however, was dependent on the magnitude of mechanical mismatch between the matrix and the cell aggregates. In general, when the elastic modulus of the matrix of the VLCM was greater than the emboli (EVLCM/Eemb > 1), the emboli were constrained to grow within the channel, and when the elastic modulus of the matrix was less than the emboli (0 < EVLCM/Eemb < 1), the emboli bulged into the matrix. Due to a large difference in myosin II expression between the cell lines, we hypothesized that tumor cell aggregate stiffness is an indicator of cellular force-generating capability. Inhibitors of myosin-related force generation decreased the elastic modulus and/or increased the stress relaxation of the tumor cell aggregates, effectively increasing the mechanical mismatch. The increased mechanical mismatch after drug treatment was correlated with increased confinement of tumor emboli growth along the channel, which may translate to increased tumor burden due to the increased tumor volume within the diffusion distance of nutrients and oxygen.
Collapse
Affiliation(s)
- Jonathan Kulwatno
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Jamie Gearhart
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Xiangyu Gong
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Nora Herzog
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Matthew Getzin
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Mihaela Skobe
- Department of Oncological Sciences & Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristen L Mills
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA.,Department of Mechanical, Aerospace, and Nuclear Engineering, Rensselaer Polytechnic Institute, Troy, NY, USA
| |
Collapse
|
178
|
Blanco‐Fernandez B, Gaspar VM, Engel E, Mano JF. Proteinaceous Hydrogels for Bioengineering Advanced 3D Tumor Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003129. [PMID: 33643799 PMCID: PMC7887602 DOI: 10.1002/advs.202003129] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/13/2020] [Indexed: 05/14/2023]
Abstract
The establishment of tumor microenvironment using biomimetic in vitro models that recapitulate key tumor hallmarks including the tumor supporting extracellular matrix (ECM) is in high demand for accelerating the discovery and preclinical validation of more effective anticancer therapeutics. To date, ECM-mimetic hydrogels have been widely explored for 3D in vitro disease modeling owing to their bioactive properties that can be further adapted to the biochemical and biophysical properties of native tumors. Gathering on this momentum, herein the current landscape of intrinsically bioactive protein and peptide hydrogels that have been employed for 3D tumor modeling are discussed. Initially, the importance of recreating such microenvironment and the main considerations for generating ECM-mimetic 3D hydrogel in vitro tumor models are showcased. A comprehensive discussion focusing protein, peptide, or hybrid ECM-mimetic platforms employed for modeling cancer cells/stroma cross-talk and for the preclinical evaluation of candidate anticancer therapies is also provided. Further development of tumor-tunable, proteinaceous or peptide 3D microtesting platforms with microenvironment-specific biophysical and biomolecular cues will contribute to better mimic the in vivo scenario, and improve the predictability of preclinical screening of generalized or personalized therapeutics.
Collapse
Affiliation(s)
- Barbara Blanco‐Fernandez
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and TechnologyBaldiri Reixac 10–12Barcelona08028Spain
| | - Vítor M. Gaspar
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| | - Elisabeth Engel
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and TechnologyBaldiri Reixac 10–12Barcelona08028Spain
- Materials Science and Metallurgical EngineeringPolytechnical University of Catalonia (UPC)Eduard Maristany 16Barcelona08019Spain
- CIBER en BioingenieríaBiomateriales y NanomedicinaCIBER‐BBNMadrid28029Spain
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials, University of AveiroCampus Universitário de SantiagoAveiro3810‐193Portugal
| |
Collapse
|
179
|
Abstract
Caveolae are bulb-like invaginations made up of two essential structural proteins, caveolin-1 and cavins, which are abundantly present at the plasma membrane of vertebrate cells. Since their discovery more than 60 years ago, the function of caveolae has been mired in controversy. The last decade has seen the characterization of new caveolae components and regulators together with the discovery of additional cellular functions that have shed new light on these enigmatic structures. Early on, caveolae and/or caveolin-1 have been involved in the regulation of several parameters associated with cancer progression such as cell migration, metastasis, angiogenesis, or cell growth. These studies have revealed that caveolin-1 and more recently cavin-1 have a dual role with either a negative or a positive effect on most of these parameters. The recent discovery that caveolae can act as mechanosensors has sparked an array of new studies that have addressed the mechanobiology of caveolae in various cellular functions. This review summarizes the current knowledge on caveolae and their role in cancer development through their activity in membrane tension buffering. We propose that the role of caveolae in cancer has to be revisited through their response to the mechanical forces encountered by cancer cells during tumor mass development.
Collapse
Affiliation(s)
- Vibha Singh
- UMR3666, INSERM U1143, Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie - Centre de Recherche, PSL Research University, CNRS, 75005, Paris, France
| | - Christophe Lamaze
- UMR3666, INSERM U1143, Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie - Centre de Recherche, PSL Research University, CNRS, 75005, Paris, France.
| |
Collapse
|
180
|
Lolo FN, Jiménez-Jiménez V, Sánchez-Álvarez M, Del Pozo MÁ. Tumor-stroma biomechanical crosstalk: a perspective on the role of caveolin-1 in tumor progression. Cancer Metastasis Rev 2021; 39:485-503. [PMID: 32514892 DOI: 10.1007/s10555-020-09900-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor stiffening is a hallmark of malignancy that actively drives tumor progression and aggressiveness. Recent research has shed light onto several molecular underpinnings of this biomechanical process, which has a reciprocal crosstalk between tumor cells, stromal fibroblasts, and extracellular matrix remodeling at its core. This dynamic communication shapes the tumor microenvironment; significantly determines disease features including therapeutic resistance, relapse, or metastasis; and potentially holds the key for novel antitumor strategies. Caveolae and their components emerge as integrators of different aspects of cell function, mechanotransduction, and ECM-cell interaction. Here, we review our current knowledge on the several pivotal roles of the essential caveolar component caveolin-1 in this multidirectional biomechanical crosstalk and highlight standing questions in the field.
Collapse
Affiliation(s)
- Fidel Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Ángel Del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
181
|
Antmen E, Demirci U, Hasirci V. Micropatterned Surfaces Expose the Coupling between Actin Cytoskeleton-Lamin/Nesprin and Nuclear Deformability of Breast Cancer Cells with Different Malignancies. Adv Biol (Weinh) 2021; 5:e2000048. [PMID: 33724728 PMCID: PMC9049775 DOI: 10.1002/adbi.202000048] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 10/16/2020] [Indexed: 12/18/2022]
Abstract
Mechanotransduction proteins transfer mechanical stimuli through nucleo-cytoskeletal coupling and affect the nuclear morphology of cancer cells. However, the contribution of actin filament integrity has never been studied directly. It is hypothesized that differences in nuclear deformability of cancer cells are influenced by the integrity of actin filaments. In this study, transparent micropatterned surfaces as simple tools to screen cytoskeletal and nuclear distortions are presented. Surfaces decorated with micropillars are used to culture and image breast cancer cells and quantify their deformation using shape descriptors (circularity, area, perimeter). Using two drugs (cytochalasin D and jasplakinolide), actin filaments are disrupted. Deformation of cells on micropillars is decreased upon drug treatment as shown by increased circularity. However, the effect is much smaller on benign MCF10A than on malignant MCF7 and MDAMB231 cells. On micropatterned surfaces, molecular analysis shows that Lamin A/C and Nesprin-2 expressions decreased but, after drug treatment, increased in malignant cells but not in benign cells. These findings suggest that Lamin A/C, Nesprin-2 and actin filaments are critical in mechanotransduction of cancer cells. Consequently, transparent micropatterned surfaces can be used as image analysis platforms to provide robust, high throughput measurements of nuclear deformability of cancer cells, including the effect of cytoskeletal elements.
Collapse
Affiliation(s)
- Ezgi Antmen
- BIOMATEN, Middle East Technical University (METU) Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey
- METU, Department of Biotechnology, Ankara, Turkey
| | - Utkan Demirci
- Department of Radiology, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Vasif Hasirci
- BIOMATEN, Middle East Technical University (METU) Center of Excellence in Biomaterials and Tissue Engineering, Ankara, Turkey
- METU, Department of Biological Sciences, Ankara, Turkey
- Acibadem Mehmet Ali Aydinlar University, Department of Medical Engineering, Atasehir, Istanbul, Turkey
| |
Collapse
|
182
|
Petersen EV, Chudakova DA, Skorova EY, Anikin V, Reshetov IV, Mynbaev OA. The Extracellular Matrix-Derived Biomarkers for Diagnosis, Prognosis, and Personalized Therapy of Malignant Tumors. Front Oncol 2020; 10:575569. [PMID: 33425730 PMCID: PMC7793707 DOI: 10.3389/fonc.2020.575569] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/10/2020] [Indexed: 01/18/2023] Open
Abstract
The tumor biomarkers already have proven clinical value and have become an integral part in cancer management and modern translational oncology. The tumor tissue microenvironment (TME), which includes extracellular matrix (ECM), signaling molecules, immune and stromal cells, and adjacent non-tumorous tissue, contributes to cancer pathogenesis. Thus, TME-derived biomarkers have many clinical applications. This review is predominately based on the most recent publications (manuscripts published in a last 5 years, or seminal publications published earlier) and fills a gap in the current literature on the cancer biomarkers derived from the TME, with particular attention given to the ECM and products of its processing and degradation, ECM-associated extracellular vesicles (EVs), biomechanical characteristics of ECM, and ECM-derived biomarkers predicting response to the immunotherapy. We discuss the clinical utility of the TME-incorporating three-dimensional in vitro and ex vivo cell culture models for personalized therapy. We conclude that ECM is a critical driver of malignancies and ECM-derived biomarkers should be included in diagnostics and prognostics panels of markers in the clinic.
Collapse
Affiliation(s)
- Elena V. Petersen
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Daria A. Chudakova
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Ekaterina Yu. Skorova
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Vladimir Anikin
- Harefield Hospital, The Royal Brompton and Harefield Hospitals NHS Foundation Trust, Harefield, United Kingdom
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Igor V. Reshetov
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Ospan A. Mynbaev
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
183
|
Modulating the Crosstalk between the Tumor and the Microenvironment Using SiRNA: A Flexible Strategy for Breast Cancer Treatment. Cancers (Basel) 2020; 12:cancers12123744. [PMID: 33322132 PMCID: PMC7763441 DOI: 10.3390/cancers12123744] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/27/2020] [Accepted: 12/04/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary With this review we aimed to collect the most relevant scientific findings regarding siRNA therapeutic tools against breast cancer microenvironment. Remarkably, breast cancer treatments have been redirected towards the tumor microenvironment components, mainly involved in patients’ relapse and pharmacological resistance. Therefore, siRNAs represent a promising strategy to jeopardize the tumor microenvironment interplay thanks to their non-toxic and specific effects. Abstract Tumorigenesis is a complex and multistep process in which sequential mutations in oncogenes and tumor-suppressor genes result in enhanced proliferation and apoptosis escape. Over the past decades, several studies have provided evidence that tumors are more than merely a mass of malignant cancer cells, with the tumor microenvironment (TME) also contributing to cancer progression. For this reason, the focus of cancer research in recent years has shifted from the malignant cancer cell itself to the TME and its interactions. Since the TME actively participates in tumor progression, therapeutic strategies targeting it have created great interest. In this context, much attention has been paid to the potential application of small interfering RNA (siRNA), a class of non-coding RNA that has the ability to downregulate the expression of target genes in a sequence-specific way. This is paving the way for a novel therapeutic approach for the treatment of several diseases, including cancer. In this review, we describe recent efforts in developing siRNA therapeutics for the treatment of breast cancer, with particular emphasis on TME regulation. We focus on studies that adapt siRNA design to reprogram/re-educate the TME and eradicate the interplay between cancer cells and TME.
Collapse
|
184
|
Mechanical strain induces phenotypic changes in breast cancer cells and promotes immunosuppression in the tumor microenvironment. J Transl Med 2020; 100:1503-1516. [PMID: 32572176 PMCID: PMC7686122 DOI: 10.1038/s41374-020-0452-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BCa) proliferates within a complex, three-dimensional microenvironment amid heterogeneous biochemical and biophysical cues. Understanding how mechanical forces within the tumor microenvironment (TME) regulate BCa phenotype is of great interest. We demonstrate that mechanical strain enhanced the proliferation and migration of both estrogen receptor+ and triple-negative (TNBC) human and mouse BCa cells. Furthermore, a critical role for exosomes derived from cells subjected to mechanical strain in these pro-tumorigenic effects was identified. Exosome production by TNBC cells increased upon exposure to oscillatory strain (OS), which correlated with elevated cell proliferation. Using a syngeneic, orthotopic mouse model of TNBC, we identified that preconditioning BCa cells with OS significantly increased tumor growth and myeloid-derived suppressor cells (MDSCs) and M2 macrophages in the TME. This pro-tumorigenic myeloid cell enrichment also correlated with a decrease in CD8+ T cells. An increase in PD-L1+ exosome release from BCa cells following OS supported additive T cell inhibitory functions in the TME. The role of exosomes in MDSC and M2 macrophage was confirmed in vivo by cytotracking fluorescent exosomes, derived from labeled 4T1.2 cells, preconditioned with OS. In addition, in vivo internalization and intratumoral localization of tumor-cell derived exosomes was observed within MDSCs, M2 macrophages, and CD45-negative cell populations following direct injection of fluorescently-labeled exosomes. Our data demonstrate that exposure to mechanical strain promotes invasive and pro-tumorigenic phenotypes in BCa cells, indicating that mechanical strain can impact the growth and proliferation of cancer cell, alter exosome production by BCa, and induce immunosuppression in the TME by dampening anti-tumor immunity.
Collapse
|
185
|
Mondal C, Di Martino JS, Bravo-Cordero JJ. Actin dynamics during tumor cell dissemination. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 360:65-98. [PMID: 33962751 PMCID: PMC8246644 DOI: 10.1016/bs.ircmb.2020.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The actin cytoskeleton is a dynamic network that regulates cellular behavior from development to disease. By rearranging the actin cytoskeleton, cells are capable of migrating and invading during developmental processes; however, many of these cellular properties are hijacked by cancer cells to escape primary tumors and disseminate to distant organs in the body. In this review article, we highlight recent work describing how cancer cells regulate the actin cytoskeleton to achieve efficient invasion and metastatic colonization. We also review new imaging technologies that are capable of revealing the complex architecture and regulation of the actin cytoskeleton during motility and invasion of tumor cells.
Collapse
Affiliation(s)
- Chandrani Mondal
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Julie S Di Martino
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Division of Hematology and Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
186
|
Richter C, Mayhew D, Rennhack JP, So J, Stover EH, Hwang JH, Szczesna-Cordary D. Genomic Amplification and Functional Dependency of the Gamma Actin Gene ACTG1 in Uterine Cancer. Int J Mol Sci 2020; 21:ijms21228690. [PMID: 33217970 PMCID: PMC7698702 DOI: 10.3390/ijms21228690] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Sarcomere and cytoskeleton genes, or actomyosin genes, regulate cell biology including mechanical stress, cell motility, and cell division. While actomyosin genes are recurrently dysregulated in cancers, their oncogenic roles have not been examined in a lineage-specific fashion. In this report, we investigated dysregulation of nine sarcomeric and cytoskeletal genes across 20 cancer lineages. We found that uterine cancers harbored the highest frequencies of amplification and overexpression of the gamma actin gene, ACTG1. Each of the four subtypes of uterine cancers, mixed endometrial carcinomas, serous carcinomas, endometroid carcinomas, and carcinosarcomas harbored between 5~20% of ACTG1 gene amplification or overexpression. Clinically, patients with ACTG1 gains had a poor prognosis. ACTG1 gains showed transcriptional patterns that reflect activation of oncogenic signals, repressed response to innate immunity, or immunotherapy. Functionally, the CRISPR-CAS9 gene deletion of ACTG1 had the most robust and consistent effects in uterine cancer cells relative to 20 other lineages. Overall, we propose that ACTG1 regulates the fitness of uterine cancer cells by modulating cell-intrinsic properties and the tumor microenvironment. In summary, the ACTG1 functions relative to other actomyosin genes support the notion that it is a potential biomarker and a target gene in uterine cancer precision therapies.
Collapse
Affiliation(s)
- Camden Richter
- Dana-Farber Cancer Institute, Harvard Medical School, Broad Institute of MIT and Harvard, Boston, MA 02215, USA; (C.R.); (D.M.); (J.P.R.); (J.S.); (E.H.S.)
| | - David Mayhew
- Dana-Farber Cancer Institute, Harvard Medical School, Broad Institute of MIT and Harvard, Boston, MA 02215, USA; (C.R.); (D.M.); (J.P.R.); (J.S.); (E.H.S.)
- Department of Radiation Oncology, Tufts Medical Center, Boston, MA 02111, USA
| | - Jonathan P. Rennhack
- Dana-Farber Cancer Institute, Harvard Medical School, Broad Institute of MIT and Harvard, Boston, MA 02215, USA; (C.R.); (D.M.); (J.P.R.); (J.S.); (E.H.S.)
| | - Jonathan So
- Dana-Farber Cancer Institute, Harvard Medical School, Broad Institute of MIT and Harvard, Boston, MA 02215, USA; (C.R.); (D.M.); (J.P.R.); (J.S.); (E.H.S.)
| | - Elizabeth H. Stover
- Dana-Farber Cancer Institute, Harvard Medical School, Broad Institute of MIT and Harvard, Boston, MA 02215, USA; (C.R.); (D.M.); (J.P.R.); (J.S.); (E.H.S.)
| | - Justin H. Hwang
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, MN 55414, USA
- Correspondence: (J.H.H.); (D.S.-C.); Tel.: +1-612-626-3003 (J.H.H.); +1-305-243-2908 (D.S.-C.); Fax: +1-612-625-6919 (J.H.H.); +1-305-243-4555 (D.S.-C.)
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, FL 33136, USA
- Correspondence: (J.H.H.); (D.S.-C.); Tel.: +1-612-626-3003 (J.H.H.); +1-305-243-2908 (D.S.-C.); Fax: +1-612-625-6919 (J.H.H.); +1-305-243-4555 (D.S.-C.)
| |
Collapse
|
187
|
Yokoyama S, Shigeishi H, Murodumi H, Sakuma M, Kato H, Higashikawa K, Ohta K, Sugiyama M, Takechi M. TGF‐β1 induces amoeboid‐to‐mesenchymal transition of CD44
high
oral squamous cell carcinoma cells via miR‐422a downregulation through ERK activation and Cofilin‐1 phosphorylation. J Oral Pathol Med 2020; 50:155-164. [DOI: 10.1111/jop.13113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 09/28/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Sho Yokoyama
- Department of Oral and Maxillofacial Surgery Program of Dentistry Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Hideo Shigeishi
- Department of Public Oral Health Program of Oral Health Sciences Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Hiroshi Murodumi
- Department of Oral and Maxillofacial Surgery Program of Dentistry Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Miyuki Sakuma
- Department of Oral and Maxillofacial Surgery Program of Dentistry Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Hiroki Kato
- Department of Oral and Maxillofacial Surgery Program of Dentistry Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Koichiro Higashikawa
- Department of Oral and Maxillofacial Surgery Program of Dentistry Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Kouji Ohta
- Department of Public Oral Health Program of Oral Health Sciences Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Masaru Sugiyama
- Department of Public Oral Health Program of Oral Health Sciences Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| | - Masaaki Takechi
- Department of Oral and Maxillofacial Surgery Program of Dentistry Graduate School of Biomedical and Health Sciences Hiroshima University Hiroshima Japan
| |
Collapse
|
188
|
Nassef MZ, Melnik D, Kopp S, Sahana J, Infanger M, Lützenberg R, Relja B, Wehland M, Grimm D, Krüger M. Breast Cancer Cells in Microgravity: New Aspects for Cancer Research. Int J Mol Sci 2020; 21:ijms21197345. [PMID: 33027908 PMCID: PMC7582256 DOI: 10.3390/ijms21197345] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/29/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is the leading cause of cancer death in females. The incidence has risen dramatically during recent decades. Dismissed as an "unsolved problem of the last century", breast cancer still represents a health burden with no effective solution identified so far. Microgravity (µg) research might be an unusual method to combat the disease, but cancer biologists decided to harness the power of µg as an exceptional method to increase efficacy and precision of future breast cancer therapies. Numerous studies have indicated that µg has a great impact on cancer cells; by influencing proliferation, survival, and migration, it shifts breast cancer cells toward a less aggressive phenotype. In addition, through the de novo generation of tumor spheroids, µg research provides a reliable in vitro 3D tumor model for preclinical cancer drug development and to study various processes of cancer progression. In summary, µg has become an important tool in understanding and influencing breast cancer biology.
Collapse
Affiliation(s)
- Mohamed Zakaria Nassef
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.Z.N.); (D.M.); (S.K.); (M.I.); (R.L.); (M.W.); (D.G.)
| | - Daniela Melnik
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.Z.N.); (D.M.); (S.K.); (M.I.); (R.L.); (M.W.); (D.G.)
| | - Sascha Kopp
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.Z.N.); (D.M.); (S.K.); (M.I.); (R.L.); (M.W.); (D.G.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Jayashree Sahana
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark;
| | - Manfred Infanger
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.Z.N.); (D.M.); (S.K.); (M.I.); (R.L.); (M.W.); (D.G.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Ronald Lützenberg
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.Z.N.); (D.M.); (S.K.); (M.I.); (R.L.); (M.W.); (D.G.)
| | - Borna Relja
- Experimental Radiology, Department of Radiology and Nuclear Medicine, Otto von Guericke University, 39120 Magdeburg, Germany;
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.Z.N.); (D.M.); (S.K.); (M.I.); (R.L.); (M.W.); (D.G.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.Z.N.); (D.M.); (S.K.); (M.I.); (R.L.); (M.W.); (D.G.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark;
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Clinic for Plastic, Aesthetic and Hand Surgery, Otto von Guericke University, 39106 Magdeburg, Germany; (M.Z.N.); (D.M.); (S.K.); (M.I.); (R.L.); (M.W.); (D.G.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-6757471
| |
Collapse
|
189
|
Hessmann E, Buchholz SM, Demir IE, Singh SK, Gress TM, Ellenrieder V, Neesse A. Microenvironmental Determinants of Pancreatic Cancer. Physiol Rev 2020; 100:1707-1751. [DOI: 10.1152/physrev.00042.2019] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) belongs to the most lethal solid tumors in humans. A histological hallmark feature of PDAC is the pronounced tumor microenvironment (TME) that dynamically evolves during tumor progression. The TME consists of different non-neoplastic cells such as cancer-associated fibroblasts, immune cells, endothelial cells, and neurons. Furthermore, abundant extracellular matrix components such as collagen and hyaluronic acid as well as matricellular proteins create a highly dynamic and hypovascular TME with multiple biochemical and physical interactions among the various cellular and acellular components that promote tumor progression and therapeutic resistance. In recent years, intensive research efforts have resulted in a significantly improved understanding of the biology and pathophysiology of the TME in PDAC, and novel stroma-targeted approaches are emerging that may help to improve the devastating prognosis of PDAC patients. However, none of anti-stromal therapies has been approved in patients so far, and there is still a large discrepancy between multiple successful preclinical results and subsequent failure in clinical trials. Furthermore, recent findings suggest that parts of the TME may also possess tumor-restraining properties rendering tailored therapies even more challenging.
Collapse
Affiliation(s)
- Elisabeth Hessmann
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Soeren M. Buchholz
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Ihsan Ekin Demir
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Shiv K. Singh
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Thomas M. Gress
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Volker Ellenrieder
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| | - Albrecht Neesse
- Department of Gastroenterology, Gastrointestinal Oncology, and Endocrinology, University Medical Centre Goettingen, Georg August University, Goettingen, Germany; Department of Surgery, Klinikum rechts der Isar, Technische Universität München, School of Medicine Munich, Munich, Germany; Sonderforschungsbereich/Collaborative Research Centre 1321 Modeling and Targeting Pancreatic Cancer, Munich, Germany; Deutsches Konsortium für Translationale Krebsforschung (DKTK) Munich Site, Munich, Germany; and
| |
Collapse
|
190
|
Bailey M, Alunni-Cardinali M, Correa N, Caponi S, Holsgrove T, Barr H, Stone N, Winlove CP, Fioretto D, Palombo F. Viscoelastic properties of biopolymer hydrogels determined by Brillouin spectroscopy: A probe of tissue micromechanics. SCIENCE ADVANCES 2020; 6:eabc1937. [PMID: 33127678 PMCID: PMC7608813 DOI: 10.1126/sciadv.abc1937] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 09/16/2020] [Indexed: 05/09/2023]
Abstract
Many problems in mechanobiology urgently require characterization of the micromechanical properties of cells and tissues. Brillouin light scattering has been proposed as an emerging optical elastography technique to meet this need. However, the information contained in the Brillouin spectrum is still a matter of debate because of fundamental problems in understanding the role of water in biomechanics and in relating the Brillouin data to low-frequency macroscopic mechanical parameters. Here, we investigate this question using gelatin as a model system in which the macroscopic physical properties can be manipulated to mimic all the relevant biological states of matter, ranging from the liquid to the gel and the glassy phase. We demonstrate that Brillouin spectroscopy is able to reveal both the elastic and viscous properties of biopolymers that are central to the structure and function of biological tissues.
Collapse
Affiliation(s)
- Michelle Bailey
- University of Exeter, School of Physics and Astronomy, Exeter EX4 4QL, UK
| | | | - Noemi Correa
- University of Exeter, School of Physics and Astronomy, Exeter EX4 4QL, UK
| | - Silvia Caponi
- CNR-IOM-Istituto Officina dei Materiali-Research Unit in Perugia, Department of Physics and Geology, University of Perugia, Perugia I-06123, Italy
| | | | - Hugh Barr
- Gloucestershire Royal Hospital, Gloucester GL1 3NN, UK
| | - Nick Stone
- University of Exeter, School of Physics and Astronomy, Exeter EX4 4QL, UK
| | - C Peter Winlove
- University of Exeter, School of Physics and Astronomy, Exeter EX4 4QL, UK
| | - Daniele Fioretto
- University of Perugia, Department of Physics and Geology, Perugia I-06123, Italy.
| | - Francesca Palombo
- University of Exeter, School of Physics and Astronomy, Exeter EX4 4QL, UK.
| |
Collapse
|
191
|
Park S, Shi Y, Kim BC, Jo MH, Cruz LO, Gou Z, Ha T, Lu LF, Reich DH, Chen Y. Force-dependent trans-endocytosis by breast cancer cells depletes costimulatory receptor CD80 and attenuates T cell activation. Biosens Bioelectron 2020; 165:112389. [PMID: 32729511 DOI: 10.1016/j.bios.2020.112389] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022]
Abstract
In this study, we investigated the biophysical interaction between cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and CD80. CTLA-4 is a key molecule in immunosuppression, and CD80 is a costimulatory receptor promoting T cell activation. We observed that after cell-cell contact was established between breast cancer cells and antigen presenting cells (APCs), CTLA-4 expressed on the breast cancer cells bind to CD80 expressed on the APCs, and underwent trans-endocytosis to deplete CD80. Force measurement and live cell imaging revealed that upon binding to CD80, forces generated by breast cancer cells and transmitted via CTLA-4 were sufficiently strong to displace CD80 from the surface of APCs to be internalized by breast cancer cells. We further demonstrated that because of the force-dependent trans-endocytosis of CD80, the capacity of APCs to activate T cells was significantly attenuated. Furthermore, inhibiting force generation in cancer cells would increase the T cell activating capacity of APCs. Our results provide a possible mechanism behind the immunosuppression commonly seen in breast cancer patients, and may lead to a new strategy to restore anti-tumor immunity by inhibiting pathways of force-generation.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical Engineering, Johns Hopkins University, MD, 21218, USA; Center for Cell Dynamics, Johns Hopkins University, MD, 21218, USA; Institute for NanoBioTechnology, Johns Hopkins University, MD, 21218, USA
| | - Yu Shi
- Department of Physics & Astronomy, Johns Hopkins University, MD, 21218, USA
| | - Byoung Choul Kim
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Howard Hughes Medical Institute, Baltimore, MD, 21205, USA; Division of Nano-Bioengineering, Incheon National University, Incheon, 22012, South Korea
| | - Myung Hyun Jo
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Leilani O Cruz
- Division of Biological Science, University of California, San Diego, CA, 92093, USA
| | - Zheming Gou
- Department of Mechanical Engineering, Johns Hopkins University, MD, 21218, USA
| | - Taekjip Ha
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Howard Hughes Medical Institute, Baltimore, MD, 21205, USA
| | - Li-Fan Lu
- Division of Biological Science, University of California, San Diego, CA, 92093, USA
| | - Daniel H Reich
- Department of Physics & Astronomy, Johns Hopkins University, MD, 21218, USA
| | - Yun Chen
- Department of Mechanical Engineering, Johns Hopkins University, MD, 21218, USA; Center for Cell Dynamics, Johns Hopkins University, MD, 21218, USA; Institute for NanoBioTechnology, Johns Hopkins University, MD, 21218, USA.
| |
Collapse
|
192
|
Tian F, Lin TC, Wang L, Chen S, Chen X, Yiu PM, Tsui OKC, Chu J, Kiang CH, Park H. Mechanical Responses of Breast Cancer Cells to Substrates of Varying Stiffness Revealed by Single-Cell Measurements. J Phys Chem Lett 2020; 11:7643-7649. [PMID: 32794712 DOI: 10.1021/acs.jpclett.0c02065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
How cancer cells respond to different mechanical environments remains elusive. Here, we investigated the tension in single focal adhesions of MDA-MB-231 (metastatic breast cancer cells) and MCF-10A (normal human breast cells) cells on substrates of varying stiffness using single-cell measurements. Tension measurements in single focal adhesions using an improved FRET-based tension sensor showed that the tension in focal adhesions of MDA-MB-231 cells increased on stiffer substrates while the tension in MCF-10A cells exhibited no apparent change against the substrate stiffness. Viscoelasticity measurements using magnetic tweezers showed that the power-law exponent of MDA-MB-231 cells decreased on stiffer substrates whereas MCF-10A cells had similar exponents throughout the whole stiffness, indicating that MDA-MB-231 cells change their viscoelasticity on stiffer substrates. Such changes in tension in focal adhesions and viscoelasticity against the substrate stiffness represent an adaptability of cancer cells in mechanical environments, which can facilitate the metastasis of cancer cells to different tissues.
Collapse
Affiliation(s)
- Fang Tian
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Tsung-Cheng Lin
- Department of Physics & Astronomy, Rice University, Houston, Texas 77005, United States
| | - Liang Wang
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Sidong Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Xingxiang Chen
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Pak Man Yiu
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Ophelia K C Tsui
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| | - Jun Chu
- Guangdong Provincial Key Laboratory of Biomedical Optical Imaging Technology & Center for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ching-Hwa Kiang
- Department of Physics & Astronomy, Rice University, Houston, Texas 77005, United States
| | - Hyokeun Park
- Department of Physics, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
- State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong
| |
Collapse
|
193
|
Ion Channels in Cancer: Orchestrators of Electrical Signaling and Cellular Crosstalk. Rev Physiol Biochem Pharmacol 2020; 183:103-133. [PMID: 32894333 DOI: 10.1007/112_2020_48] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ion channels are pore-forming transmembrane proteins that govern ion flux to regulate a myriad of biological processes in development, physiology, and disease. Across various types of cancer, ion channel expression and activity are often dysregulated. We review the contribution of ion channels to multiple stages of tumorigenesis based on data from in vivo model systems. As intertumoral and intratumoral heterogeneities are major obstacles in developing effective therapies, we provide perspectives on how ion channels in tumor cells and their microenvironment represent targetable vulnerabilities in the areas of tumor-stromal cell interactions, cancer neuroscience, and cancer mechanobiology.
Collapse
|
194
|
Heimer S, Knoll G, Neubert P, Hammer KP, Wagner S, Bauer RJ, Jantsch J, Ehrenschwender M. Hypertonicity counteracts MCL-1 and renders BCL-XL a synthetic lethal target in head and neck cancer. FEBS J 2020; 288:1822-1838. [PMID: 32710568 DOI: 10.1111/febs.15492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 06/09/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an aggressive and difficult-to-treat cancer entity. Current therapies ultimately aim to activate the mitochondria-controlled (intrinsic) apoptosis pathway, but complex alterations in intracellular signaling cascades and the extracellular microenvironment hamper treatment response. On the one hand, proteins of the BCL-2 family set the threshold for cell death induction and prevent accidental cellular suicide. On the other hand, controlling a cell's readiness to die also determines whether malignant cells are sensitive or resistant to anticancer treatments. Here, we show that HNSCC cells upregulate the proapoptotic BH3-only protein NOXA in response to hyperosmotic stress. Induction of NOXA is sufficient to counteract the antiapoptotic properties of MCL-1 and switches HNSCC cells from dual BCL-XL/MCL-1 protection to exclusive BCL-XL addiction. Hypertonicity-induced functional loss of MCL-1 renders BCL-XL a synthetically lethal target in HNSCC, and inhibition of BCL-XL efficiently kills HNSCC cells that poorly respond to conventional therapies. We identify hypertonicity-induced upregulation of NOXA as link between osmotic pressure in the tumor environment and mitochondrial priming, which could perspectively be exploited to boost efficacy of anticancer drugs.
Collapse
Affiliation(s)
- Sina Heimer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Gertrud Knoll
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Patrick Neubert
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Karin P Hammer
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Richard J Bauer
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany.,Department of Oral and Maxillofacial Surgery, Center for Medical Biotechnology, University Hospital Regensburg, Regensburg, Germany
| | - Jonathan Jantsch
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Martin Ehrenschwender
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
195
|
Characterizing the ecological and evolutionary dynamics of cancer. Nat Genet 2020; 52:759-767. [DOI: 10.1038/s41588-020-0668-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
|
196
|
D'Angelo E, Lindoso RS, Sensi F, Pucciarelli S, Bussolati B, Agostini M, Collino F. Intrinsic and Extrinsic Modulators of the Epithelial to Mesenchymal Transition: Driving the Fate of Tumor Microenvironment. Front Oncol 2020; 10:1122. [PMID: 32793478 PMCID: PMC7393251 DOI: 10.3389/fonc.2020.01122] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022] Open
Abstract
The epithelial to mesenchymal transition (EMT) is an evolutionarily conserved process. In cancer, EMT can activate biochemical changes in tumor cells that enable the destruction of the cellular polarity, leading to the acquisition of invasive capabilities. EMT regulation can be triggered by intrinsic and extrinsic signaling, allowing the tumor to adapt to the microenvironment demand in the different stages of tumor progression. In concomitance, tumor cells undergoing EMT actively interact with the surrounding tumor microenvironment (TME) constituted by cell components and extracellular matrix as well as cell secretome elements. As a result, the TME is in turn modulated by the EMT process toward an aggressive behavior. The current review presents the intrinsic and extrinsic modulators of EMT and their relationship with the TME, focusing on the non-cell-derived components, such as secreted metabolites, extracellular matrix, as well as extracellular vesicles. Moreover, we explore how these modulators can be suitable targets for anticancer therapy and personalized medicine.
Collapse
Affiliation(s)
- Edoardo D'Angelo
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- LIFELAB Program, Consorzio per la Ricerca Sanitaria–CORIS, Veneto Region, Padua, Italy
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
| | - Rafael Soares Lindoso
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine–REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Francesca Sensi
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
- Department of Molecular Sciences and Nanosystems, Cà Foscari University of Venice, Venice, Italy
| | - Salvatore Pucciarelli
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
| | - Benedetta Bussolati
- Department of Medical Sciences, Molecular Biotechnology Center, University of Torino, Turin, Italy
| | - Marco Agostini
- First Surgical Clinic, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- LIFELAB Program, Consorzio per la Ricerca Sanitaria–CORIS, Veneto Region, Padua, Italy
- Institute of Pediatric Research, Fondazione Citta della Speranza, Padua, Italy
| | - Federica Collino
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione Ca' Granda, IRCCS Policlinico di Milano, Milan, Italy
| |
Collapse
|
197
|
Asem M, Young A, Oyama C, ClaureDeLaZerda A, Liu Y, Ravosa MJ, Gupta V, Jewell A, Khabele D, Stack MS. Ascites-induced compression alters the peritoneal microenvironment and promotes metastatic success in ovarian cancer. Sci Rep 2020; 10:11913. [PMID: 32681052 PMCID: PMC7367827 DOI: 10.1038/s41598-020-68639-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 06/26/2020] [Indexed: 12/17/2022] Open
Abstract
The majority of women with recurrent ovarian cancer (OvCa) develop malignant ascites with volumes that can reach > 2 L. The resulting elevation in intraperitoneal pressure (IPP), from normal values of 5 mmHg to as high as 22 mmHg, causes striking changes in the loading environment in the peritoneal cavity. The effect of ascites-induced changes in IPP on OvCa progression is largely unknown. Herein we model the functional consequences of ascites-induced compression on ovarian tumor cells and components of the peritoneal microenvironment using a panel of in vitro, ex vivo and in vivo assays. Results show that OvCa cell adhesion to the peritoneum was increased under compression. Moreover, compressive loads stimulated remodeling of peritoneal mesothelial cell surface ultrastructure via induction of tunneling nanotubes (TNT). TNT-mediated interaction between peritoneal mesothelial cells and OvCa cells was enhanced under compression and was accompanied by transport of mitochondria from mesothelial cells to OvCa cells. Additionally, peritoneal collagen fibers adopted a more linear anisotropic alignment under compression, a collagen signature commonly correlated with enhanced invasion in solid tumors. Collectively, these findings elucidate a new role for ascites-induced compression in promoting metastatic OvCa progression.
Collapse
Affiliation(s)
- Marwa Asem
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Allison Young
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Carlysa Oyama
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Alejandro ClaureDeLaZerda
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Yueying Liu
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Matthew J Ravosa
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Vijayalaxmi Gupta
- Department of Obstetrics & Gynecology, Medical Center, University of Kansas, Kansas City, USA
| | - Andrea Jewell
- Department of Obstetrics & Gynecology, Medical Center, University of Kansas, Kansas City, USA
| | - Dineo Khabele
- Department of Obstetrics & Gynecology, Medical Center, University of Kansas, Kansas City, USA
| | - M Sharon Stack
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA.
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA.
| |
Collapse
|
198
|
Characterization of multicellular breast tumor spheroids using image data-driven biophysical mathematical modeling. Sci Rep 2020; 10:11583. [PMID: 32665565 PMCID: PMC7360601 DOI: 10.1038/s41598-020-68324-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
Multicellular tumor spheroid (MCTS) systems provide an in vitro cell culture model system which mimics many of the complexities of an in vivo solid tumor and tumor microenvironment, and are often used to study cancer cell growth and drug efficacy. Here, we present a coupled experimental-computational framework to estimate phenotypic growth and biophysical tumor microenvironment properties. This novel framework utilizes standard microscopy imaging of MCTS systems to drive a biophysical mathematical model of MCTS growth and mechanical interactions. By extending our previous in vivo mechanically-coupled reaction–diffusion modeling framework we developed a microscopy image processing framework capable of mechanistic characterization of MCTS systems. Using MDA-MB-231 breast cancer MCTS, we estimated biophysical parameters of cellular diffusion, rate of cellular proliferation, and cellular tractions forces. We found significant differences in these model-based biophysical parameters throughout the treatment time course between untreated and treated MCTS systems, whereas traditional size-based morphometric parameters were inconclusive. The proposed experimental-computational framework estimates mechanistic MCTS growth and invasion parameters with significant potential to assist in better and more precise assessment of in vitro drug efficacy through the development of computational analysis methodologies for three-dimensional cell culture systems to improve the development and evaluation of antineoplastic drugs.
Collapse
|
199
|
Zhang D, Li C, Huang Z. Relaxation time constant based optical coherence elastography. JOURNAL OF BIOPHOTONICS 2020; 13:e201960233. [PMID: 32166913 DOI: 10.1002/jbio.201960233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/19/2020] [Accepted: 03/08/2020] [Indexed: 06/10/2023]
Abstract
This study aimed at visualizing relative relaxation time constant (RTC) in soft tissue by using optical coherence elastography (OCE). We proposed a forced vibration model as a theoretical base to express RTC using axial gradient of periodic vibration phase captured by phase sensitive optical coherence tomography (PhS-OCT). Validation of the model had been accomplished by experiments with isotropic and double-layered phantoms. A fresh chicken breast sample treated with focused ultrasound was prepared to test performance of the RTC-OCE in real tissue. All results were cross-validated with indentation test and traditional strain-based elastography. This study first utilized RTC mapping in 2D and 3D that covers the information of both elasticity and viscosity. The generated RTC mapping revealed the same mechanical difference internal sample which is correlated with conventional strain mapping. RTC mapping is potentially to be served as new biomarker for disease diagnosis in the future.
Collapse
Affiliation(s)
- Duo Zhang
- School of Science and Engineering, University of Dundee, Dundee, Scotland, UK
| | - Chunhui Li
- School of Science and Engineering, University of Dundee, Dundee, Scotland, UK
| | - Zhihong Huang
- School of Science and Engineering, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
200
|
MacDonald L, Jenkins J, Purvis G, Lee J, Franco AT. The Thyroid Tumor Microenvironment: Potential Targets for Therapeutic Intervention and Prognostication. Discov Oncol 2020; 11:205-217. [PMID: 32548798 DOI: 10.1007/s12672-020-00390-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022] Open
Abstract
Thyroid cancer is the most common endocrine malignancy and incidences are rising rapidly, in both pediatric and adult populations. Many thyroid tumors are successfully treated which results in low mortality rates, but there is often a significant morbidity associated with thyroid cancer treatments. For patients with tumors that are not successfully treated with surgical resection or radioactive iodine treatment, prognosis is dramatically reduced. Patients diagnosed with anaplastic thyroid cancer face a very grim prognosis with a median survival of 6 months post-diagnosis. There is a critical need to identify patients who are at greatest risk of developing persistent disease and progressing to poorly differentiated or anaplastic disease. Furthermore, development of treatments associated with less morbidity would represent a significant improvement for thyroid cancer survivors. It is well established the stromal cells and components of the tumor microenvironment can drive tumor progression and resistance to therapy. Here we review the current state of what is known regarding the thyroid tumor microenvironment and how these factors may contribute to thyroid tumor pathogenesis. Study of the tumor microenvironment within thyroid cancer is a relatively new field, and more studies are needed to dissect the complex and dynamic crosstalk between thyroid tumor cells and its tumor niche.
Collapse
Affiliation(s)
| | | | - Grace Purvis
- Division of Endocrinology and Diabetes Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Joshua Lee
- Division of Endocrinology and Diabetes Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aime T Franco
- Division of Endocrinology and Diabetes Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|