51
|
Roles of DDX5 in the tumorigenesis, proliferation, differentiation, metastasis and pathway regulation of human malignancies. Biochim Biophys Acta Rev Cancer 2019; 1871:85-98. [DOI: 10.1016/j.bbcan.2018.11.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 02/07/2023]
|
52
|
Merchant B, Edelstein-Keshet L, Feng JJ. A Rho-GTPase based model explains spontaneous collective migration of neural crest cell clusters. Dev Biol 2018; 444 Suppl 1:S262-S273. [DOI: 10.1016/j.ydbio.2018.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023]
|
53
|
Nobre AR, Entenberg D, Wang Y, Condeelis J, Aguirre-Ghiso JA. The Different Routes to Metastasis via Hypoxia-Regulated Programs. Trends Cell Biol 2018; 28:941-956. [PMID: 30041830 PMCID: PMC6214449 DOI: 10.1016/j.tcb.2018.06.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/28/2018] [Accepted: 06/29/2018] [Indexed: 12/18/2022]
Abstract
Hypoxia is linked to metastasis; however, how it affects metastatic progression is not clear due to limited consensus in the literature. We posit that this lack of consensus is due to hypoxia being studied using different approaches, such as in vitro, primary tumor, or metastasis assays in an isolated manner. Here, we review the pros and cons of in vitro hypoxia assays, highlight in vivo studies that inform on physiological hypoxia, and review the evidence that primary tumor hypoxia might influence the fate of disseminated tumor cells (DTCs) in secondary organs. Our analysis suggests that consensus can be reached by using in vivo methods of study, which also allow better modeling of how hypoxia affects DTC fate and metastasis.
Collapse
Affiliation(s)
- Ana Rita Nobre
- Division of Hematology and Oncology, Department of Medicine, Department of Otolaryngology, Department of Oncological Sciences, Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA; Abel Salazar School of Biomedicine, Porto University, Porto, Portugal; These authors contributed equally
| | - David Entenberg
- Department of Anatomy and Structural Biology, Gruss Lipper Biophotonics Center, Integrated Imaging Program, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA; These authors contributed equally
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Gruss Lipper Biophotonics Center, Integrated Imaging Program, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA
| | - John Condeelis
- Department of Anatomy and Structural Biology, Gruss Lipper Biophotonics Center, Integrated Imaging Program, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY 10461, USA.
| | - Julio A Aguirre-Ghiso
- Division of Hematology and Oncology, Department of Medicine, Department of Otolaryngology, Department of Oncological Sciences, Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY 10029, USA.
| |
Collapse
|
54
|
Li R, Serrano JC, Xing H, Lee TA, Azizgolshani H, Zaman M, Kamm RD. Interstitial flow promotes macrophage polarization toward an M2 phenotype. Mol Biol Cell 2018; 29:1927-1940. [PMID: 29995595 PMCID: PMC6232969 DOI: 10.1091/mbc.e18-03-0164] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tumor tissues are characterized by an elevated interstitial fluid flow from the tumor to the surrounding stroma. Macrophages in the tumor microenvironment are key contributors to tumor progression. While it is well established that chemical stimuli within the tumor tissues can alter macrophage behaviors, the effects of mechanical stimuli, especially the flow of interstitial fluid in the tumor microenvironment, on macrophage phenotypes have not been explored. Here, we used three-dimensional biomimetic models to reveal that macrophages can sense and respond to pathophysiological levels of interstitial fluid flow reported in tumors (∼3 µm/s). Specifically, interstitial flow (IF) polarizes macrophages toward an M2-like phenotype via integrin/Src-mediated mechanotransduction pathways involving STAT3/6. Consistent with this flow-induced M2 polarization, macrophages treated with IF migrate faster and have an enhanced ability to promote cancer cell migration. Moreover, IF directs macrophages to migrate against the flow. Since IF emanates from the tumor to the surrounding stromal tissues, our results suggest that IF could not only induce M2 polarization of macrophages but also recruit these M2 macrophages toward the tumor masses, contributing to cancer cell invasion and tumor progression. Collectively, our study reveals that IF could be a critical regulator of tumor immune environment.
Collapse
Affiliation(s)
- Ran Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jean Carlos Serrano
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Hao Xing
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Tara A Lee
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Hesham Azizgolshani
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Muhammad Zaman
- Department of Biomedical Engineering, Boston University, Boston, MA 02215
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
55
|
hMENA isoforms impact NSCLC patient outcome through fibronectin/β1 integrin axis. Oncogene 2018; 37:5605-5617. [PMID: 29907768 PMCID: PMC6193944 DOI: 10.1038/s41388-018-0364-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 05/23/2018] [Accepted: 05/23/2018] [Indexed: 12/22/2022]
Abstract
We demonstrated previously that the splicing of the actin regulator, hMENA, generates two alternatively expressed isoforms, hMENA11a and hMENAΔv6, which have opposite functions in cell invasiveness. Their mechanisms of action have remained unclear. Here we report two major findings: (i) hMENA regulates β1 integrin expression. This was shown by depleting total hMENA, which led to loss of nuclear expression of serum response factor (SRF)-coactivator myocardin-related transcription factor 1 (MRTF-A), leading to an increase in the G-actin/F-actin ratio crucial for MRTF-A localization. This in turn inhibited SRF activity and the expression of its target gene β1 integrin. (ii) hMENA11a reduces and hMENAΔv6 increases β1 integrin activation and signaling. Moreover, exogenous expression of hMENA11a in hMENAΔv6-positive cancer cells dramatically reduces secretion of extracellular matrix (ECM) components, including β1 integrin ligands and metalloproteinases. On the other hand, overexpression of the pro-invasive hMENAΔv6 increases fibronectin production. In primary tumors high hMENA11a correlates with low stromal fibronectin and a favorable clinical outcome of early node-negative non-small-cell lung cancer patients. These data provide new insights into the roles of hMENA11a and hMENAΔv6 in the druggable β1 integrin-ECM signaling axis and allow stratification of patient risk, guiding their clinical management.
Collapse
|
56
|
Meirson T, Gil-Henn H. Targeting invadopodia for blocking breast cancer metastasis. Drug Resist Updat 2018; 39:1-17. [PMID: 30075834 DOI: 10.1016/j.drup.2018.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/04/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
Dissemination of cancer cells from the primary tumor and their spread to distant sites of the body is the leading cause of mortality in metastatic cancer patients. Metastatic cancer cells invade surrounding tissues and blood vessels by forming F-actin-rich protrusions known as invadopodia, which degrade the extracellular matrix and enable invasion of tumor cells through it. Invadopodia have now been observed in vivo, and recent evidence demonstrates direct molecular links between assembly of invadopodia and cancer metastasis in both mouse models and in human patients. While significant progress has been achieved in the last decade in understanding the molecular mechanisms and signaling pathways regulating invadopodia formation and function, the application of this knowledge to development of prognostic and therapeutic approaches for cancer metastasis has not been discussed before. Here, we provide a detailed overview of current prognostic markers and tests for cancer metastasis and discuss their advantages, disadvantages, and their predicted efficiency. Using bioinformatic patient database analysis, we demonstrate, for the first time, a significant correlation between invadopodia-associated genes to breast cancer metastasis, suggesting that invadopodia could be used as both a prognostic marker and as a therapeutic target for blocking cancer metastasis. We include here a novel network interaction map of invadopodia-associated proteins with currently available inhibitors, demonstrating a central role for the recently identified EGFR-Pyk2-Src-Arg-cortactin invadopodial pathway, to which re-purposing of existent inhibitors could be used to block breast cancer metastasis. We then present an updated overview of current cancer-related clinical trials, demonstrating the negligible number of trials focusing on cancer metastasis. We also discuss the difficulties and complexity of performing cancer metastasis clinical trials, and the possible development of anti-metastasis drug resistance when using a prolonged preventive treatment with invadopodia inhibitors. This review presents a new perspective on invadopodia-mediated tumor invasiveness and may lead to the development of novel prognostic and therapeutic approaches for cancer metastasis.
Collapse
Affiliation(s)
- Tomer Meirson
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel.
| |
Collapse
|
57
|
Arwert EN, Harney AS, Entenberg D, Wang Y, Sahai E, Pollard JW, Condeelis JS. A Unidirectional Transition from Migratory to Perivascular Macrophage Is Required for Tumor Cell Intravasation. Cell Rep 2018; 23:1239-1248. [PMID: 29719241 PMCID: PMC5946803 DOI: 10.1016/j.celrep.2018.04.007] [Citation(s) in RCA: 185] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/12/2018] [Accepted: 03/30/2018] [Indexed: 12/30/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are critical for tumor metastasis. Two TAM subsets support cancer cell intravasation: migratory macrophages guide cancer cells toward blood vessels, where sessile perivascular macrophages assist their entry into the blood. However, little is known about the inter-relationship between these functionally distinct TAMs or their possible inter-conversion. We show that motile, streaming TAMs are newly arrived monocytes, recruited via CCR2 signaling, that then differentiate into the sessile perivascular macrophages. This unidirectional process is regulated by CXCL12 and CXCR4. Cancer cells induce TGF-β-dependent upregulation of CXCR4 in monocytes, while CXCL12 expressed by perivascular fibroblasts attracts these motile TAMs toward the blood vessels, bringing motile cancer cells with them. Once on the blood vessel, the migratory TAMs differentiate into perivascular macrophages, promoting vascular leakiness and intravasation.
Collapse
Affiliation(s)
- Esther N Arwert
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK; Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA
| | - Allison S Harney
- Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA
| | - David Entenberg
- Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA
| | - Yarong Wang
- Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA
| | - Erik Sahai
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK.
| | - Jeffrey W Pollard
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, NY 10461, USA.
| | - John S Condeelis
- Gruss-Lipper Biophotonics Center and the Integrated Imaging Program, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
58
|
Intravital microscopy in the study of the tumor microenvironment: from bench to human application. Oncotarget 2018; 9:20165-20178. [PMID: 29732011 PMCID: PMC5929454 DOI: 10.18632/oncotarget.24957] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/15/2018] [Indexed: 12/31/2022] Open
Abstract
Intravital microscopy (IVM) is a dynamic imaging modality that allows for the real time observation of biologic processes in vivo, including angiogenesis and immune cell interactions. In the setting of preclinical cancer models, IVM has facilitated an understanding of the tumor associated vasculature and the role of effector immune cells in the tumor microenvironment. Novel approaches to apply IVM to human malignancies have thus far focused on cancer diagnosis and tumor vessel characterization, but have the potential to provide advances in the field of personalized medicine by identifying individual patients who may respond to systemically delivered chemotherapeutic drugs or immunotherapeutic agents. In this review, we highlight the role that IVM has had in investigating tumor vasculature and the tumor microenvironment in preclinical studies and discuss its current and future applications to directly observe human tumors.
Collapse
|
59
|
Li Y, Liu TM. Discovering Macrophage Functions Using In Vivo Optical Imaging Techniques. Front Immunol 2018; 9:502. [PMID: 29599778 PMCID: PMC5863475 DOI: 10.3389/fimmu.2018.00502] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/26/2018] [Indexed: 12/27/2022] Open
Abstract
Macrophages are an important component of host defense and inflammation and play a pivotal role in immune regulation, tissue remodeling, and metabolic regulation. Since macrophages are ubiquitous in human bodies and have versatile physiological functions, they are involved in virtually every disease, including cancer, diabetes, multiple sclerosis, and atherosclerosis. Molecular biological and histological methods have provided critical information on macrophage biology. However, many in vivo dynamic behaviors of macrophages are poorly understood and yet to be discovered. A better understanding of macrophage functions and dynamics in pathogenesis will open new opportunities for better diagnosis, prognostic assessment, and therapeutic intervention. In this article, we will review the advances in macrophage tracking and analysis with in vivo optical imaging in the context of different diseases. Moreover, this review will cover the challenges and solutions for optical imaging techniques during macrophage intravital imaging.
Collapse
Affiliation(s)
- Yue Li
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Tzu-Ming Liu
- Faculty of Health Sciences, University of Macau, Macao, China
| |
Collapse
|
60
|
Chemotherapy-induced metastasis: mechanisms and translational opportunities. Clin Exp Metastasis 2018; 35:269-284. [PMID: 29307118 DOI: 10.1007/s10585-017-9870-x] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
Tumors often overcome the cytotoxic effects of chemotherapy through either acquired or environment-mediated drug resistance. In addition, signals from the microenvironment obfuscate the beneficial effects of chemotherapy and may facilitate progression and metastatic dissemination. Seminal mediators in chemotherapy-induced metastasis appear to be a wide range of hematopoietic, mesenchymal and immune progenitor cells, originating from the bone marrow. The actual purpose of these cells is to orchestrate the repair response to the cytotoxic damage of chemotherapy. However, these repair responses are exploited by tumor cells at every step of the metastatic cascade, ranging from tumor cell invasion, intravasation and hematogenous dissemination to extravasation and effective colonization at the metastatic site. A better understanding of the mechanistic underpinnings of chemotherapy-induced metastasis will allow us to better predict which patients are more likely to exhibit pro-metastatic responses to chemotherapy and will help develop new therapeutic strategies to neutralize chemotherapy-driven prometastatic changes.
Collapse
|
61
|
Roh-Johnson M, Shah AN, Stonick JA, Poudel KR, Kargl J, Yang GH, di Martino J, Hernandez RE, Gast CE, Zarour LR, Antoku S, Houghton AM, Bravo-Cordero JJ, Wong MH, Condeelis J, Moens CB. Macrophage-Dependent Cytoplasmic Transfer during Melanoma Invasion In Vivo. Dev Cell 2018; 43:549-562.e6. [PMID: 29207258 DOI: 10.1016/j.devcel.2017.11.003] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/15/2017] [Accepted: 11/04/2017] [Indexed: 12/31/2022]
Abstract
Interactions between tumor cells and tumor-associated macrophages play critical roles in the initiation of tumor cell motility. To capture the cellular interactions of the tumor microenvironment with high-resolution imaging, we directly visualized tumor cells and their interactions with macrophages in zebrafish. Live imaging in zebrafish revealed that macrophages are dynamic, yet maintain sustained contact with tumor cells. In addition, the recruitment of macrophages to tumor cells promotes tumor cell dissemination. Using a Cre/LoxP strategy, we found that macrophages transfer cytoplasm to tumor cells in zebrafish and mouse models. Remarkably, macrophage cytoplasmic transfer correlated with melanoma cell dissemination. We further found that macrophages transfer cytoplasm to tumor cells upon cell contact in vitro. Thus, we present a model in which macrophage/tumor cell contact allows for the transfer of cytoplasmic molecules from macrophages to tumor cells corresponding to increased tumor cell motility and dissemination.
Collapse
Affiliation(s)
- Minna Roh-Johnson
- Fred Hutchinson Cancer Research Center, Basic Sciences Division, Seattle, WA 98109, USA.
| | - Arish N Shah
- Fred Hutchinson Cancer Research Center, Basic Sciences Division, Seattle, WA 98109, USA
| | - Jason A Stonick
- Fred Hutchinson Cancer Research Center, Basic Sciences Division, Seattle, WA 98109, USA
| | - Kumud R Poudel
- Fred Hutchinson Cancer Research Center, Basic Sciences Division, Seattle, WA 98109, USA
| | - Julia Kargl
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA 98109, USA; Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz 8036, Austria
| | - Grace H Yang
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA 98109, USA
| | - Julie di Martino
- Icahn School of Medicine at Mount Sinai, Division of Hematology and Oncology, Department of Medicine, New York, NY 10029, USA
| | | | - Charles E Gast
- Oregon Health & Science University, Department of Cell, Developmental, and Cancer Biology, The Knight Cancer Institute, Portland, OR 97239, USA
| | - Luai R Zarour
- Oregon Health & Science University, Department of Surgery, Portland, OR 97239, USA
| | - Susumu Antoku
- Columbia University, Department of Pathology and Cell Biology, New York, NY 10027, USA
| | - A McGarry Houghton
- Fred Hutchinson Cancer Research Center, Clinical Research Division, Seattle, WA 98109, USA
| | - Jose Javier Bravo-Cordero
- Icahn School of Medicine at Mount Sinai, Division of Hematology and Oncology, Department of Medicine, New York, NY 10029, USA
| | - Melissa H Wong
- Oregon Health & Science University, Department of Cell, Developmental, and Cancer Biology, The Knight Cancer Institute, Portland, OR 97239, USA
| | - John Condeelis
- Albert Einstein College of Medicine, Department of Anatomy and Structural Biology, Bronx, NY 10461, USA
| | - Cecilia B Moens
- Fred Hutchinson Cancer Research Center, Basic Sciences Division, Seattle, WA 98109, USA
| |
Collapse
|
62
|
Zhang B, Cao M, He Y, Liu Y, Zhang G, Yang C, Du Y, Xu J, Hu J, Gao F. Combination of plasma HA and circulating M2-like monocytes may serve as a diagnostic marker for breast cancer. J Cancer 2017; 8:3522-3530. [PMID: 29151937 PMCID: PMC5687167 DOI: 10.7150/jca.20227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/28/2017] [Indexed: 12/23/2022] Open
Abstract
Background: Breast cancer (BC)-derived hyaluronan (HA) can induce the formation of M2-like tumor-associated macrophages (TAMs) in tumor context. However, little is known about the correlation between circulating M2-like monocytes and plasma HA in BC patients. This study focused on evaluating the relationship between circulating M2-like monocytes and plasma HA, and further appraised the diagnostic value of them in BC. Methods: The expression of M2-like TAMs and HA was determined in pathological tissues by immunohistochemistry. Flow cytometry was used to detect the levels of circulating CD14+CD204+ M2-like monocytes in 81 BC patients, 45 patients with breast benign diseases, and 46 healthy subjects. The levels of HA, CEA, and CA15-3 were measured in plasma samples using chemiluminescence method. Results: M2-like TAMs and HA expressions were elevated in BC tissues compared with benign tissues. In correspondence, the frequency of circulating CD14+CD204+ M2-like monocytes and the plasma HA levels were significantly higher in patients with BC than those in control groups. Importantly, there was a positive correlation between circulating M2-like monocytes and the plasma HA (Spearman r = 0.404, p < 0.001). Area under receiver operating characteristic curve (ROC) for the combination of circulating M2-like monocytes and HA was 0.899 (95% CI: 0.853-0.946), which was higher than the panel of CEA and CA15-3. Conclusions: The frequency of circulating CD14+CD204+ M2-like monocytes was positively correlated to plasma HA levels. The combination of circulating CD14+CD204+ M2-like monocytes and plasma HA could provide considerable diagnostic value in BC.
Collapse
Affiliation(s)
- Boke Zhang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Manlin Cao
- Department of Rehabilitation Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Yiqing He
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Yiwen Liu
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Guoliang Zhang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Cuixia Yang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Yan Du
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Jing Xu
- Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Jiajie Hu
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| | - Feng Gao
- Department of Molecular Biology and Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, PR China
| |
Collapse
|
63
|
Mak M, Anderson S, McDonough MC, Spill F, Kim JE, Boussommier-Calleja A, Zaman MH, Kamm RD. Integrated Analysis of Intracellular Dynamics of MenaINV Cancer Cells in a 3D Matrix. Biophys J 2017; 112:1874-1884. [PMID: 28494958 DOI: 10.1016/j.bpj.2017.03.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/20/2017] [Accepted: 03/27/2017] [Indexed: 01/17/2023] Open
Abstract
The intracellular environment is composed of a filamentous network that exhibits dynamic turnover of cytoskeletal components and internal force generation from molecular motors. Particle tracking microrheology enables a means to probe the internal mechanics and dynamics. Here, we develop an analytical model to capture the basic features of the active intracellular mechanical environment, including both thermal and motor-driven effects, and show consistency with a diverse range of experimental microrheology data. We further perform microrheology experiments, integrated with Brownian dynamics simulations of the active cytoskeleton, on metastatic breast cancer cells embedded in a three-dimensional collagen matrix with and without the presence of epidermal growth factor to probe the intracellular mechanical response in a physiologically mimicking scenario. Our results demonstrate that EGF stimulation can alter intracellular stiffness and power output from molecular motor-driven fluctuations in cells overexpressing an invasive isoform of the actin-associated protein Mena.
Collapse
Affiliation(s)
- Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut; Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts; Department of Biomedical Engineering, Boston University, Boston, Massachusetts.
| | | | - Meghan C McDonough
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Fabian Spill
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts; Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Jessica E Kim
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | | | - Muhammad H Zaman
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts; Howard Hughes Medical Institute, Boston University, Boston, Massachusetts.
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.
| |
Collapse
|
64
|
Prognostic values of cancer associated macrophage-like cells (CAML) enumeration in metastatic breast cancer. Breast Cancer Res Treat 2017; 165:733-741. [PMID: 28687903 DOI: 10.1007/s10549-017-4372-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/30/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Circulating cancer associated macrophage-like cells (CAMLs) have been detected in the peripheral blood of patients with solid tumors including breast cancer. However, the prognostic relevance of CAMLs in metastatic breast cancer (MBC) has not been evaluated. In the present study, we aimed to measure CAMLs and circulating tumor cells (CTCs) at baseline and examine their prognostic value in patients with MBC. METHODS Peripheral blood samples from 127 MBC patients were collected at baseline before starting a new treatment. The detection and enumeration of CAMLs and CTCs in 7.5 ml whole blood were performed using the CellSearch™ system. The associations of CAMLs and CTCs with the progression-free survival (PFS) and overall survival (OS) in the patients were evaluated using Kaplan-Meier curves and Cox proportional hazards modeling. RESULTS Among 127 MBC patients, 21 (16.5%) were detected with CAMLs and 38 (29.9%) had elevated CTCs (≥5 CTCs/7.5 ml). Patients with CAMLs at baseline had worse PFS and OS with an adjusted hazard ratio (HR) of 1.75 (95% CI 1.03-2.98, P = 0.0374) and 3.75 (95% CI 1.52-9.26, P = 0.0042), compared to patients without CAMLs. Compared with patients with <5 CTCs and without CAMLs, patients with <5 CTCs and with CAMLs, with ≥5 CTCs but without CAMLs, or with ≥5 CTCs and with CAMLs, had an increasing trend of risk of disease progression (HR = 0.84, 3.42 and 4.04 respectively, P for trend <0.0001) and death (HR = 2.66, 6.14, and 9.13, respectively, P for trend <0.0001). CONCLUSION Baseline enumeration of individual CAMLs is an independent indicator for MBC patients' survival. Evaluation of CAMLs in peripheral blood might provide a potential biomarker with additional prognostic values over CTC enumeration alone in MBC patients.
Collapse
|
65
|
Karagiannis GS, Pastoriza JM, Wang Y, Harney AS, Entenberg D, Pignatelli J, Sharma VP, Xue EA, Cheng E, D'Alfonso TM, Jones JG, Anampa J, Rohan TE, Sparano JA, Condeelis JS, Oktay MH. Neoadjuvant chemotherapy induces breast cancer metastasis through a TMEM-mediated mechanism. Sci Transl Med 2017; 9:eaan0026. [PMID: 28679654 PMCID: PMC5592784 DOI: 10.1126/scitranslmed.aan0026] [Citation(s) in RCA: 359] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/13/2017] [Indexed: 12/11/2022]
Abstract
Breast cancer cells disseminate through TIE2/MENACalc/MENAINV-dependent cancer cell intravasation sites, called tumor microenvironment of metastasis (TMEM), which are clinically validated as prognostic markers of metastasis in breast cancer patients. Using fixed tissue and intravital imaging of a PyMT murine model and patient-derived xenografts, we show that chemotherapy increases the density and activity of TMEM sites and Mena expression and promotes distant metastasis. Moreover, in the residual breast cancers of patients treated with neoadjuvant paclitaxel after doxorubicin plus cyclophosphamide, TMEM score and its mechanistically connected MENAINV isoform expression pattern were both increased, suggesting that chemotherapy, despite decreasing tumor size, increases the risk of metastatic dissemination. Chemotherapy-induced TMEM activity and cancer cell dissemination were reversed by either administration of the TIE2 inhibitor rebastinib or knockdown of the MENA gene. Our results indicate that TMEM score increases and MENA isoform expression pattern changes with chemotherapy and can be used in predicting prometastatic changes in response to chemotherapy. Furthermore, inhibitors of TMEM function may improve clinical benefits of chemotherapy in the neoadjuvant setting or in metastatic disease.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jessica M Pastoriza
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Yarong Wang
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Allison S Harney
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Radiology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeanine Pignatelli
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ved P Sharma
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emily A Xue
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Esther Cheng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Timothy M D'Alfonso
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Joan G Jones
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jesus Anampa
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joseph A Sparano
- Department of Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
| |
Collapse
|
66
|
Differences in Three-Dimensional Geometric Recognition by Non-Cancerous and Cancerous Epithelial Cells on Microgroove-Based Topography. Sci Rep 2017; 7:4244. [PMID: 28652607 PMCID: PMC5484713 DOI: 10.1038/s41598-017-03779-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 05/04/2017] [Indexed: 01/08/2023] Open
Abstract
During metastasis, cancer cells are exposed to various three-dimensional microstructures within the body, but the relationship between cancer migration and three-dimensional geometry remain largely unclear. Here, such geometric effects on cancerous cells were investigated by characterizing the motility of various cancer cell types on microgroove-based topographies made of polydimethylsiloxane (PDMS), with particular emphasis on distinguishing cancerous and non-cancerous epithelial cells, as well as understanding the underlying mechanism behind such differences. The 90-degree walls enhanced motility for all cell lines, but the degrees of enhancements were less pronounced for the cancerous cells. Interestingly, while the non-cancerous epithelial cell types conformed to the three-dimensional geometrical cues and migrated along the walls, the cancerous cell types exhibited a unique behavior of climbing upright walls, and this was associated with the inability to form stable, polarized actin cytoskeleton along the walls of the microgrooves. Furthermore, when non-cancerous epithelial cell lines were altered to different levels of polarization capabilities and cancer malignancy or treated with inhibitory drugs, their three-dimensional geometry-dependent motility approached those of cancerous cell lines. Overall, the results suggest that cancerous cells may gradually lose geometrical recognition with increasing cancer malignancy, allowing them to roam freely ignoring three-dimensional geometrical cues during metastasis.
Collapse
|
67
|
Zhang B, Cao M, He Y, Liu Y, Zhang G, Yang C, Du Y, Xu J, Hu J, Gao F. Increased circulating M2-like monocytes in patients with breast cancer. Tumour Biol 2017. [PMID: 28639912 DOI: 10.1177/1010428317711571] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Boke Zhang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Manlin Cao
- Department of Rehabilitation Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Yiqing He
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Yiwen Liu
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Guoliang Zhang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Cuixia Yang
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Yan Du
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Jing Xu
- Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Jiajie Hu
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| | - Feng Gao
- Department of Molecular Biology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
- Department of Clinical Laboratory, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, P.R. China
| |
Collapse
|
68
|
Milano DF, Natividad RJ, Saito Y, Luo CY, Muthuswamy SK, Asthagiri AR. Positive Quantitative Relationship between EMT and Contact-Initiated Sliding on Fiber-like Tracks. Biophys J 2017; 111:1569-1574. [PMID: 27705778 DOI: 10.1016/j.bpj.2016.08.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/28/2016] [Accepted: 08/26/2016] [Indexed: 12/31/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a complex process by which cells acquire invasive properties that enable escape from the primary tumor. Complete EMT, however, is not required for metastasis: circulating tumor cells exhibit hybrid epithelial-mesenchymal states, and genetic perturbations promoting partial EMT induce metastasis in vivo. An open question is whether and to what extent intermediate stages of EMT promote invasiveness. Here, we investigate this question, building on recent observation of a new invasive property. Migrating cancer cell lines and cells transduced with prometastatic genes slide around other cells on spatially confined, fiberlike micropatterns. We show here that low-dosage/short-duration exposure to transforming growth factor beta (TGFβ) induces partial EMT and enables sliding on narrower (26 μm) micropatterns than untreated counterparts (41 μm). High-dosage/long-duration exposure induces more complete EMT, including disrupted cell-cell contacts and reduced E-cadherin expression, and promotes sliding on the narrowest (15 μm) micropatterns. These results identify a direct and quantitative relationship between EMT and cell sliding and show that EMT-associated invasive sliding is progressive, with cells that undergo partial EMT exhibiting intermediate sliding behavior and cells that transition more completely through EMT displaying maximal sliding. Our findings suggest a model in which fiber maturation and EMT work synergistically to promote invasiveness during cancer progression.
Collapse
Affiliation(s)
- Daniel F Milano
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Robert J Natividad
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Yasuhiro Saito
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Catherine Y Luo
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Senthil K Muthuswamy
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Anand R Asthagiri
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts; Department of Bioengineering, Northeastern University, Boston, Massachusetts; Department of Biology, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
69
|
Cadherin composition and multicellular aggregate invasion in organotypic models of epithelial ovarian cancer intraperitoneal metastasis. Oncogene 2017. [PMID: 28628116 PMCID: PMC5648607 DOI: 10.1038/onc.2017.171] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During epithelial ovarian cancer (EOC) progression, intraperitoneally disseminating tumor cells and multi-cellular aggregates (MCAs) present in ascites fluid adhere to the peritoneum and induce retraction of the peritoneal mesothelial monolayer prior to invasion of the collagen-rich sub-mesothelial matrix and proliferation into macro-metastases. Clinical studies have shown heterogeneity among EOC metastatic units with respect to cadherin expression profiles and invasive behavior, however the impact of distinct cadherin profiles on peritoneal anchoring of metastatic lesions remains poorly understood. In the current study, we demonstrate that metastasis-associated behaviors of ovarian cancer cells and MCAs are influenced by cellular cadherin composition. Our results show that mesenchymal N-cadherin expressing (Ncad+) cells and MCAs invade much more efficiently than E-cadherin expressing (Ecad+) cells. Ncad+ MCAs exhibit rapid lateral dispersal prior to penetration of three-dimensional collagen matrices. When seeded as individual cells, lateral migration and cell-cell junction formation precede matrix invasion. Neutralizing the Ncad extracellular domain with the monoclonal antibody GC-4 suppresses lateral dispersal and cell penetration of collagen gels. In contrast, use of a broad spectrum matrix metalloproteinase (MMP) inhibitor (GM6001) to block endogenous membrane type 1 matrix metalloproteinase (MT1-MMP) activity does not fully inhibit cell invasion. Using intact tissue explants, Ncad+ MCAs were also shown to efficiently rupture peritoneal mesothelial cells, exposing the sub-mesothelial collagen matrix. Acquisition of Ncad by E-cadherin expressing cells (Ecad+) increased mesothelial clearance activity, but was not sufficient to induce matrix invasion. Furthermore, co-culture of Ncad+ with Ecad+ cells did not promote a “leader-follower” mode of collective cell invasion, demonstrating that matrix remodeling and creation of invasive micro-tracks are not sufficient for cell penetration of collagen matrices in the absence of Ncad. Collectively, our data emphasize the role of Ncad in intraperitoneal seeding of EOC and provide the rationale for future studies targeting Ncad+ in pre-clinical models of EOC metastasis.
Collapse
|
70
|
Dwyer AR, Greenland EL, Pixley FJ. Promotion of Tumor Invasion by Tumor-Associated Macrophages: The Role of CSF-1-Activated Phosphatidylinositol 3 Kinase and Src Family Kinase Motility Signaling. Cancers (Basel) 2017; 9:E68. [PMID: 28629162 PMCID: PMC5483887 DOI: 10.3390/cancers9060068] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/08/2017] [Accepted: 06/12/2017] [Indexed: 12/12/2022] Open
Abstract
Macrophages interact with cells in every organ to facilitate tissue development, function and repair. However, the close interaction between macrophages and parenchymal cells can be subverted in disease, particularly cancer. Motility is an essential capacity for macrophages to be able to carry out their various roles. In cancers, the macrophage's interstitial migratory ability is frequently co-opted by tumor cells to enable escape from the primary tumor and metastatic spread. Macrophage accumulation within and movement through a tumor is often stimulated by tumor cell production of the mononuclear phagocytic growth factor, colony-stimulating factor-1 (CSF-1). CSF-1 also regulates macrophage survival, proliferation and differentiation, and its many effects are transduced by its receptor, the CSF-1R, via phosphotyrosine motif-activated signals. Mutational analysis of CSF-1R signaling indicates that the major mediators of CSF-1-induced motility are phosphatidyl-inositol-3 kinase (PI3K) and one or more Src family kinase (SFK), which activate signals to adhesion, actin polymerization, polarization and, ultimately, migration and invasion in macrophages. The macrophage transcriptome, including that of the motility machinery, is very complex and highly responsive to the environment, with selective expression of proteins and splice variants rarely found in other cell types. Thus, their unique motility machinery can be specifically targeted to block macrophage migration, and thereby, inhibit tumor invasion and metastasis.
Collapse
Affiliation(s)
- Amy R Dwyer
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Eloise L Greenland
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| | - Fiona J Pixley
- School of Biomedical Sciences, The University of Western Australia, 35 Stirling Highway, Crawley, WA 6009, Australia.
| |
Collapse
|
71
|
Milano DF, Ngai NA, Muthuswamy SK, Asthagiri AR. Regulators of Metastasis Modulate the Migratory Response to Cell Contact under Spatial Confinement. Biophys J 2017; 110:1886-1895. [PMID: 27119647 DOI: 10.1016/j.bpj.2016.02.040] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/19/2016] [Accepted: 02/23/2016] [Indexed: 11/16/2022] Open
Abstract
The breast tumor microenvironment (TMEN) is a unique niche where protein fibers help to promote invasion and metastasis. Cells migrating along these fibers are constantly interacting with each other. How cells respond to these interactions has important implications. Cancer cells that circumnavigate or slide around other cells on protein fibers take a less tortuous path out of the primary tumor; conversely, cells that turn back upon encountering other cells invade less efficiently. The contact response of migrating cancer cells in a fibrillar TMEN is poorly understood. Here, using high-aspect ratio micropatterns as a model fibrillar platform, we show that metastatic cells overcome spatial constraints to slide effectively on narrow fiber-like dimensions, whereas nontransformed MCF-10A mammary epithelial cells require much wider micropatterns to achieve moderate levels of sliding. Downregulating the cell-cell adhesion protein, E-cadherin, enables MCF-10A cells to slide on narrower micropatterns; meanwhile, introducing exogenous E-cadherin in metastatic MDA-MB-231 cells increases the micropattern dimension at which they slide. We propose the characteristic fibrillar dimension (CFD) at which effective sliding is achieved as a metric of sliding ability under spatial confinement. Using this metric, we show that metastasis-promoting genetic perturbations enhance cell sliding and reduce CFD. Activation of ErbB2 combined with downregulation of the tumor suppressor and cell polarity regulator, PARD3, reduced the CFD, in agreement with their cooperative role in inducing metastasis in vivo. The CFD was further reduced by a combination of ErbB2 activation and transforming growth factor β stimulation, which is known to enhance invasive behavior. These findings demonstrate that sliding is a quantitative property and a decrease in CFD is an effective metric to understand how multiple genetic hits interact to change cell behavior in fibrillar environments. This quantitative framework sheds insights into how genetic perturbations conspire with fibrillar maturation in the TMEN to drive the invasive behavior of cancer cells.
Collapse
Affiliation(s)
- Daniel F Milano
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts
| | - Nicholas A Ngai
- Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Senthil K Muthuswamy
- Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada; Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, Massachusetts.
| | - Anand R Asthagiri
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts; Department of Bioengineering, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
72
|
Suijkerbuijk SJE, van Rheenen J. From good to bad: Intravital imaging of the hijack of physiological processes by cancer cells. Dev Biol 2017; 428:328-337. [PMID: 28473106 DOI: 10.1016/j.ydbio.2017.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
Abstract
Homeostasis of tissues is tightly regulated at the cellular, tissue and organismal level. Interestingly, tumor cells have found ways to hijack many of these physiological processes at all the different levels. Here we review how intravital microscopy techniques have provided new insights into our understanding of tissue homeostasis and cancer progression. In addition, we highlight the different strategies that tumor cells have adopted to use these physiological processes for their own benefit. We describe how visualization of these dynamic processes in living mice has broadened to our view on cancer initiation and progression.
Collapse
Affiliation(s)
- Saskia J E Suijkerbuijk
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands
| | - Jacco van Rheenen
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
73
|
Tumor Cell Invadopodia: Invasive Protrusions that Orchestrate Metastasis. Trends Cell Biol 2017; 27:595-607. [PMID: 28412099 DOI: 10.1016/j.tcb.2017.03.003] [Citation(s) in RCA: 270] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 12/26/2022]
Abstract
Invadopodia are a subset of invadosomes that are implicated in the integration of signals from the tumor microenvironment to support tumor cell invasion and dissemination. Recent progress has begun to define how tumor cells regulate the plasticity necessary for invadopodia to assemble and function efficiently in the different microenvironments encountered during dissemination in vivo. Exquisite mapping by many laboratories of the pathways involved in integrating diverse invadopodium initiation signals, from growth factors, to extracellular matrix (ECM) and cell-cell contact in the tumor microenvironment, has led to insight into the molecular basis of this plasticity. Here, we integrate this new information to discuss how the invadopodium is an important conductor that orchestrates tumor cell dissemination during metastasis.
Collapse
|
74
|
Mekhdjian AH, Kai F, Rubashkin MG, Prahl LS, Przybyla LM, McGregor AL, Bell ES, Barnes JM, DuFort CC, Ou G, Chang AC, Cassereau L, Tan SJ, Pickup MW, Lakins JN, Ye X, Davidson MW, Lammerding J, Odde DJ, Dunn AR, Weaver VM. Integrin-mediated traction force enhances paxillin molecular associations and adhesion dynamics that increase the invasiveness of tumor cells into a three-dimensional extracellular matrix. Mol Biol Cell 2017; 28:1467-1488. [PMID: 28381423 PMCID: PMC5449147 DOI: 10.1091/mbc.e16-09-0654] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 03/24/2017] [Accepted: 03/29/2017] [Indexed: 12/21/2022] Open
Abstract
Mammary tumor cells adopt a basal-like phenotype when invading through a dense, stiffened, 3D matrix. These cells exert higher integrin-mediated traction forces, consistent with a physical motor-clutch model, display an altered molecular organization at the nanoscale, and recruit a suite of paxillin-associated proteins implicated in metastasis. Metastasis requires tumor cells to navigate through a stiff stroma and squeeze through confined microenvironments. Whether tumors exploit unique biophysical properties to metastasize remains unclear. Data show that invading mammary tumor cells, when cultured in a stiffened three-dimensional extracellular matrix that recapitulates the primary tumor stroma, adopt a basal-like phenotype. Metastatic tumor cells and basal-like tumor cells exert higher integrin-mediated traction forces at the bulk and molecular levels, consistent with a motor-clutch model in which motors and clutches are both increased. Basal-like nonmalignant mammary epithelial cells also display an altered integrin adhesion molecular organization at the nanoscale and recruit a suite of paxillin-associated proteins implicated in invasion and metastasis. Phosphorylation of paxillin by Src family kinases, which regulates adhesion turnover, is similarly enhanced in the metastatic and basal-like tumor cells, fostered by a stiff matrix, and critical for tumor cell invasion in our assays. Bioinformatics reveals an unappreciated relationship between Src kinases, paxillin, and survival of breast cancer patients. Thus adoption of the basal-like adhesion phenotype may favor the recruitment of molecules that facilitate tumor metastasis to integrin-based adhesions. Analysis of the physical properties of tumor cells and integrin adhesion composition in biopsies may be predictive of patient outcome.
Collapse
Affiliation(s)
- Armen H Mekhdjian
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | - FuiBoon Kai
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Matthew G Rubashkin
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Louis S Prahl
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455
| | - Laralynne M Przybyla
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Alexandra L McGregor
- Nancy E. and Peter C. Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - Emily S Bell
- Nancy E. and Peter C. Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - J Matthew Barnes
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Christopher C DuFort
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Guanqing Ou
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Alice C Chang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | - Luke Cassereau
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Steven J Tan
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | - Michael W Pickup
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Jonathan N Lakins
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143
| | - Xin Ye
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142
| | - Michael W Davidson
- National High Magnetic Field Laboratory and Department of Biological Science, Florida State University, Tallahassee, FL 32306
| | - Jan Lammerding
- Nancy E. and Peter C. Meinig School of Biomedical Engineering and Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853
| | - David J Odde
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455
| | - Alexander R Dunn
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143 .,Departments of Anatomy, Bioengineering and Therapeutic Sciences, and Radiation Oncology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, and UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
75
|
Santiago-Medina M, Yang J. MENA Promotes Tumor-Intrinsic Metastasis through ECM Remodeling and Haptotaxis. Cancer Discov 2017; 6:474-6. [PMID: 27138561 DOI: 10.1158/2159-8290.cd-16-0231] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Oudin and colleagues report a novel and specific function of MENA in mediating directional migration of breast cancer cells toward a fibronectin gradient of increasing concentration. This MENA-mediated haptotactic response depends on the binding of MENA to the α5β1 integrin receptor, adhesion protein signaling, and fibronectin fibrillogenesis. Cancer Discov; 6(5); 474-6. ©2016 AACRSee related article by Oudin et al., p. 516.
Collapse
Affiliation(s)
- Miguel Santiago-Medina
- Department of Pharmacology, University of California, San Diego, La Jolla, California. Moores Cancer Center, University of California, San Diego, La Jolla, California.
| | - Jing Yang
- Department of Pharmacology, University of California, San Diego, La Jolla, California. Moores Cancer Center, University of California, San Diego, La Jolla, California. Department of Pediatrics, University of California, San Diego, La Jolla, California.
| |
Collapse
|
76
|
Sparano JA, Gray R, Oktay MH, Entenberg D, Rohan T, Xue X, Donovan M, Peterson M, Shuber A, Hamilton DA, D’Alfonso T, Goldstein LJ, Gertler F, Davidson NE, Condeelis J, Jones J. A metastasis biomarker (MetaSite Breast™ Score) is associated with distant recurrence in hormone receptor-positive, HER2-negative early-stage breast cancer. NPJ Breast Cancer 2017; 3:42. [PMID: 29138761 PMCID: PMC5678158 DOI: 10.1038/s41523-017-0043-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 08/24/2017] [Accepted: 09/22/2017] [Indexed: 12/16/2022] Open
Abstract
Metastasis is the primary cause of death in early-stage breast cancer. We evaluated the association between a metastasis biomarker, which we call "Tumor Microenviroment of Metastasis" (TMEM), and risk of recurrence. TMEM are microanatomic structures where invasive tumor cells are in direct contact with endothelial cells and macrophages, and which serve as intravasation sites for tumor cells into the circulation. We evaluated primary tumors from 600 patients with Stage I-III breast cancer treated with adjuvant chemotherapy in trial E2197 (NCT00003519), plus endocrine therapy for hormone receptor (HR)+ disease. TMEM were identified and enumerated using an analytically validated, fully automated digital pathology/image analysis method (MetaSite Breast™), hereafter referred to as MetaSite Score (MS). The objectives were to determine the association between MS and distant relapse free interval (DRFI) and relapse free interval (RFI). MS was not associated with tumor size or nodal status, and correlated poorly with Oncotype DX Recurrence Score (r = 0.29) in 297 patients with HR+/HER2- disease. Proportional hazards models revealed a significant positive association between continuous MS and DRFI (p = 0.001) and RFI (p = 0.00006) in HR+/HER2- disease in years 0-5, and by MS tertiles for DRFI (p = 0.04) and RFI (p = 0.01), but not after year 5 or in triple negative or HER2+ disease. Multivariate models in HR+/HER- disease including continuous MS, clinical covariates, and categorical Recurrence Score (<18, 18-30, > 30) showed MS is an independent predictor for 5-year RFI (p = 0.05). MetaSite Score provides prognostic information for early recurrence complementary to clinicopathologic features and Recurrence Score.
Collapse
Affiliation(s)
- Joseph A. Sparano
- 0000 0001 2152 0791grid.240283.fMontefiore Medical Center, Albert Einstein College of Medicine, 1695 Eastchester Road, 10461 Bronx, NY USA
| | | | - Maja H. Oktay
- 0000 0001 2152 0791grid.240283.fMontefiore Medical Center, Albert Einstein College of Medicine, 1695 Eastchester Road, 10461 Bronx, NY USA
| | - David Entenberg
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| | - Thomas Rohan
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| | - Xiaonan Xue
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| | - Michael Donovan
- 0000 0001 0670 2351grid.59734.3cMt. Sinai School of Medicine, New York, NY USA
| | | | | | | | | | - Lori J. Goldstein
- 0000 0004 0456 6466grid.412530.1Fox Chase Cancer Center, Philadelphia, PA USA
| | - Frank Gertler
- 0000 0001 2341 2786grid.116068.8Massachusetts Institute of Technology, Boston, MA USA
| | - Nancy E. Davidson
- 0000 0004 0456 9819grid.478063.eUniversity of Pittsburgh Cancer Institute, Pittsburgh, PA USA
| | - John Condeelis
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| | - Joan Jones
- 0000 0001 2152 0791grid.240283.fAlbert Einstein College of Medicine, Bronx, NY USA
| |
Collapse
|
77
|
Pandya P, Orgaz JL, Sanz-Moreno V. Modes of invasion during tumour dissemination. Mol Oncol 2016; 11:5-27. [PMID: 28085224 PMCID: PMC5423224 DOI: 10.1002/1878-0261.12019] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/24/2016] [Accepted: 10/28/2016] [Indexed: 02/06/2023] Open
Abstract
Cancer cell migration and invasion underlie metastatic dissemination, one of the major problems in cancer. Tumour cells exhibit a striking variety of invasion strategies. Importantly, cancer cells can switch between invasion modes in order to cope with challenging environments. This ability to switch migratory modes or plasticity highlights the challenges behind antimetastasis therapy design. In this Review, we present current knowledge on different tumour invasion strategies, the determinants controlling plasticity and arising therapeutic opportunities. We propose that targeting master regulators controlling plasticity is needed to hinder tumour dissemination and metastasis.
Collapse
Affiliation(s)
- Pahini Pandya
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Jose L Orgaz
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Victoria Sanz-Moreno
- Tumour Plasticity Team, Randall Division of Cell and Molecular Biophysics, King's College London, UK
| |
Collapse
|
78
|
Pignatelli J, Bravo-Cordero JJ, Roh-Johnson M, Gandhi SJ, Wang Y, Chen X, Eddy RJ, Xue A, Singer RH, Hodgson L, Oktay MH, Condeelis JS. Macrophage-dependent tumor cell transendothelial migration is mediated by Notch1/Mena INV-initiated invadopodium formation. Sci Rep 2016; 6:37874. [PMID: 27901093 PMCID: PMC5129016 DOI: 10.1038/srep37874] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/02/2016] [Indexed: 12/27/2022] Open
Abstract
The process of intravasation involving transendothelial migration is a key step in metastatic spread. How the triple cell complex composed of a macrophage, Mena over-expressing tumor cell and endothelial cell, called the tumor microenvironment of metastasis (TMEM), facilitates tumor cell transendothelial migration is not completely understood. Previous work has shown that the physical contact between a macrophage and tumor cell results in the formation of invadopodia, actin-rich matrix degrading protrusions, important for tumor cell invasion and transendothelial migration and tumor cell dissemination. Herein, we show that the macrophage-induced invadopodium is formed through a Notch1/MenaINV signaling pathway in the tumor cell upon macrophage contact. This heterotypic tumor cell – macrophage interaction results in the upregulation of MenaINV through the activation of MENA transcription. Notch1 and MenaINV expression are required for tumor cell transendothelial migration, a necessary step during intravasation. Inhibition of the Notch signaling pathway blocked macrophage-induced invadopodium formation in vitro and the dissemination of tumor cells from the primary tumor in vivo. Our findings indicate a novel role for Notch1 signaling in the regulation of MenaINV expression and transendothelial migration and provide mechanistic information essential to the use of therapeutic inhibitors of metastasis.
Collapse
Affiliation(s)
- Jeanine Pignatelli
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Jose Javier Bravo-Cordero
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Minna Roh-Johnson
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Saumil J Gandhi
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Yarong Wang
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Xiaoming Chen
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Robert J Eddy
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Alice Xue
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Robert H Singer
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Louis Hodgson
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - Maja H Oktay
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Department of Pathology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| | - John S Condeelis
- Department of Anatomy and Structural Biology Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States.,Gruss Lipper Biophotonics Center Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, United States
| |
Collapse
|
79
|
Blood vessel endothelium-directed tumor cell streaming in breast tumors requires the HGF/C-Met signaling pathway. Oncogene 2016; 36:2680-2692. [PMID: 27893712 PMCID: PMC5426963 DOI: 10.1038/onc.2016.421] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 09/13/2016] [Accepted: 09/30/2016] [Indexed: 01/14/2023]
Abstract
During metastasis to distant sites, tumor cells migrate to blood vessels. In vivo, breast tumor cells utilize a specialized mode of migration known as streaming, where a linear assembly of tumor cells migrate directionally towards blood vessels on fibronectin-collagen I-containing extracellular matrix (ECM) fibers in response to chemotactic signals. We have successfully reconstructed tumor cell streaming in vitro by co-plating tumors cells, macrophages and endothelial cells on 2.5 μm thick ECM-coated micro-patterned substrates. We found that tumor cells and macrophages, when plated together on the micro-patterned substrates, do not demonstrate sustained directional migration in only one direction (sustained directionality) but show random bi-directional walking. Sustained directionality of tumor cells as seen in vivo was established in vitro when beads coated with human umbilical vein endothelial cells were placed at one end of the micro-patterned 'ECM fibers' within the assay. We demonstrated that these endothelial cells supply the hepatocyte growth factor (HGF) required for the chemotactic gradient responsible for sustained directionality. Using this in vitro reconstituted streaming system, we found that directional streaming is dependent on, and most effectively blocked, by inhibiting the HGF/C-Met signaling pathway between endothelial cells and tumor cells. Key observations made with the in vitro reconstituted system implicating C-Met signaling were confirmed in vivo in mammary tumors using the in vivo invasion assay and intravital multiphoton imaging of tumor cell streaming. These results establish HGF/C-Met as a central organizing signal in blood vessel-directed tumor cell migration in vivo and highlight a promising role for C-Met inhibitors in blocking tumor cell streaming and metastasis in vivo, and for use in human trials.
Collapse
|
80
|
Ellies LG. Collagen and fibronectin: threads linking obesity and breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:S50. [PMID: 27868018 PMCID: PMC5104618 DOI: 10.21037/atm.2016.10.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Lesley G Ellies
- Department of Pathology, UC San Diego, La Jolla, CA 92093, USA; Moores Cancer Center, UC San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
81
|
Mena INV dysregulates cortactin phosphorylation to promote invadopodium maturation. Sci Rep 2016; 6:36142. [PMID: 27824079 PMCID: PMC5099927 DOI: 10.1038/srep36142] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/11/2016] [Indexed: 01/12/2023] Open
Abstract
Invadopodia, actin-based protrusions of invasive carcinoma cells that focally activate extracellular matrix-degrading proteases, are essential for the migration and intravasation of tumor cells during dissemination from the primary tumor. We have previously shown that cortactin phosphorylation at tyrosine residues, in particular tyrosine 421, promotes actin polymerization at newly-forming invadopodia, promoting their maturation to matrix-degrading structures. However, the mechanism by which cells regulate the cortactin tyrosine phosphorylation-dephosphorylation cycle at invadopodia is unknown. Mena, an actin barbed-end capping protein antagonist, is expressed as various splice-isoforms. The MenaINV isoform is upregulated in migratory and invasive sub-populations of breast carcinoma cells, and is involved in tumor cell intravasation. Here we show that forced MenaINV expression increases invadopodium maturation to a far greater extent than equivalent expression of other Mena isoforms. MenaINV is recruited to invadopodium precursors just after their initial assembly at the plasma membrane, and promotes the phosphorylation of cortactin tyrosine 421 at invadopodia. In addition, we show that cortactin phosphorylation at tyrosine 421 is suppressed by the phosphatase PTP1B, and that PTP1B localization to the invadopodium is reduced by MenaINV expression. We conclude that MenaINV promotes invadopodium maturation by inhibiting normal dephosphorylation of cortactin at tyrosine 421 by the phosphatase PTP1B.
Collapse
|
82
|
Oudin MJ, Barbier L, Schäfer C, Kosciuk T, Miller MA, Han S, Jonas O, Lauffenburger DA, Gertler FB. MENA Confers Resistance to Paclitaxel in Triple-Negative Breast Cancer. Mol Cancer Ther 2016; 16:143-155. [PMID: 27811011 DOI: 10.1158/1535-7163.mct-16-0413] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/24/2016] [Accepted: 10/26/2016] [Indexed: 12/17/2022]
Abstract
Taxane therapy remains the standard of care for triple-negative breast cancer. However, high frequencies of recurrence and progression in treated patients indicate that metastatic breast cancer cells can acquire resistance to this drug. The actin regulatory protein MENA and particularly its invasive isoform, MENAINV, are established drivers of metastasis. MENAINV expression is significantly correlated with metastasis and poor outcome in human patients with breast cancer. We investigated whether MENA isoforms might play a role in driving resistance to chemotherapeutics. We find that both MENA and MENAINV confer resistance to the taxane paclitaxel, but not to the widely used DNA-damaging agents doxorubicin or cisplatin. Furthermore, paclitaxel treatment does not attenuate growth of MENAINV-driven metastatic lesions. Mechanistically, MENA isoform expression alters the ratio of dynamic and stable microtubule populations in paclitaxel-treated cells. MENA expression also increases MAPK signaling in response to paclitaxel treatment. Decreasing ERK phosphorylation by co-treatment with MEK inhibitor restored paclitaxel sensitivity by driving microtubule stabilization in MENA isoform-expressing cells. Our results reveal a novel mechanism of taxane resistance in highly metastatic breast cancer cells and identify a combination therapy to overcome such resistance. Mol Cancer Ther; 16(1); 143-55. ©2016 AACR.
Collapse
Affiliation(s)
- Madeleine J Oudin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Lucie Barbier
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,ENS-Cachan, Cachan, France
| | - Claudia Schäfer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Tatsiana Kosciuk
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Miles A Miller
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Sangyoon Han
- Lydia Hill Department for Bioinformatics, UT Southwestern Medical Center, Dallas, Texas
| | - Oliver Jonas
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Douglas A Lauffenburger
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Frank B Gertler
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts. .,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
83
|
Balsamo M, Mondal C, Carmona G, McClain LM, Riquelme DN, Tadros J, Ma D, Vasile E, Condeelis JS, Lauffenburger DA, Gertler FB. The alternatively-included 11a sequence modifies the effects of Mena on actin cytoskeletal organization and cell behavior. Sci Rep 2016; 6:35298. [PMID: 27748415 PMCID: PMC5066228 DOI: 10.1038/srep35298] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/27/2016] [Indexed: 11/09/2022] Open
Abstract
During tumor progression, alternative splicing gives rise to different Mena protein isoforms. We analyzed how Mena11a, an isoform enriched in epithelia and epithelial-like cells, affects Mena-dependent regulation of actin dynamics and cell behavior. While other Mena isoforms promote actin polymerization and drive membrane protrusion, we find that Mena11a decreases actin polymerization and growth factor-stimulated membrane protrusion at lamellipodia. Ectopic Mena11a expression slows mesenchymal-like cell motility, while isoform-specific depletion of endogenous Mena11a in epithelial-like tumor cells perturbs cell:cell junctions and increases membrane protrusion and overall cell motility. Mena11a can dampen membrane protrusion and reduce actin polymerization in the absence of other Mena isoforms, indicating that it is not simply an inactive Mena isoform. We identify a phosphorylation site within 11a that is required for some Mena11a-specific functions. RNA-seq data analysis from patient cohorts demonstrates that the difference between mRNAs encoding constitutive Mena sequences and those containing the 11a exon correlates with metastasis in colorectal cancer, suggesting that 11a exon exclusion contributes to invasive phenotypes and leads to poor clinical outcomes.
Collapse
Affiliation(s)
- Michele Balsamo
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chandrani Mondal
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Guillaume Carmona
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leslie M McClain
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daisy N Riquelme
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jenny Tadros
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Duan Ma
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Eliza Vasile
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Frank B Gertler
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
84
|
Martin OA, Anderson RL, Narayan K, MacManus MP. Does the mobilization of circulating tumour cells during cancer therapy cause metastasis? Nat Rev Clin Oncol 2016; 14:32-44. [PMID: 27550857 DOI: 10.1038/nrclinonc.2016.128] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite progressive improvements in the management of patients with locoregionally confined, advanced-stage solid tumours, distant metastasis remains a very common - and usually fatal - mode of failure after attempted curative treatment. Surgery and radiotherapy are the primary curative modalities for these patients, often combined with each other and/or with chemotherapy. Distant metastasis occurring after treatment can arise from previously undetected micrometastases or, alternatively, from persistent locoregional disease. Another possibility is that treatment itself might sometimes cause or promote metastasis. Surgical interventions in patients with cancer, including biopsies, are commonly associated with increased concentrations of circulating tumour cells (CTCs). High CTC numbers are associated with an unfavourable prognosis in many cancers. Radiotherapy and systemic antitumour therapies might also mobilize CTCs. We review the preclinical and clinical data concerning cancer treatments, CTC mobilization and other factors that might promote metastasis. Contemporary treatment regimens represent the best available curative options for patients who might otherwise die from locally confined, advanced-stage cancers; however, if such treatments can promote metastasis, this process must be understood and addressed therapeutically to improve patient survival.
Collapse
Affiliation(s)
- Olga A Martin
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Grattan street, Melbourne, Victoria 3000, Australia
| | - Robin L Anderson
- Metastasis Research Laboratory, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Grattan street, Melbourne, Victoria 3000, Australia
| | - Kailash Narayan
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Grattan street, Melbourne, Victoria 3000, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Grattan street, Melbourne, Victoria 3000, Australia
| | - Michael P MacManus
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, Victoria 3000, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Grattan street, Melbourne, Victoria 3000, Australia
| |
Collapse
|
85
|
Harney AS, Wang Y, Condeelis JS, Entenberg D. Extended Time-lapse Intravital Imaging of Real-time Multicellular Dynamics in the Tumor Microenvironment. J Vis Exp 2016. [PMID: 27341448 DOI: 10.3791/54042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
In the tumor microenvironment, host stromal cells interact with tumor cells to promote tumor progression, angiogenesis, tumor cell dissemination and metastasis. Multicellular interactions in the tumor microenvironment can lead to transient events including directional tumor cell motility and vascular permeability. Quantification of tumor vascular permeability has frequently used end-point experiments to measure extravasation of vascular dyes. However, due to the transient nature of multicellular interactions and vascular permeability, the kinetics of these dynamic events cannot be discerned. By labeling cells and vasculature with injectable dyes or fluorescent proteins, high-resolution time-lapse intravital microscopy has allowed the direct, real-time visualization of transient events in the tumor microenvironment. Here we describe a method for using multiphoton microscopy to perform extended intravital imaging in live mice to directly visualize multicellular dynamics in the tumor microenvironment. This method details cellular labeling strategies, the surgical preparation of a mammary skin flap, the administration of injectable dyes or proteins by tail vein catheter and the acquisition of time-lapse images. The time-lapse sequences obtained from this method facilitate the visualization and quantitation of the kinetics of cellular events of motility and vascular permeability in the tumor microenvironment.
Collapse
Affiliation(s)
- Allison S Harney
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Department of Radiology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine; Integrated Imaging Program, Albert Einstein College of Medicine;
| | - Yarong Wang
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine
| | - John S Condeelis
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine; Integrated Imaging Program, Albert Einstein College of Medicine
| | - David Entenberg
- Department of Anatomy & Structural Biology, Albert Einstein College of Medicine; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine; Integrated Imaging Program, Albert Einstein College of Medicine
| |
Collapse
|
86
|
Wang Y, Wang H, Li J, Entenberg D, Xue A, Wang W, Condeelis J. Direct visualization of the phenotype of hypoxic tumor cells at single cell resolution in vivo using a new hypoxia probe. INTRAVITAL 2016; 5. [PMID: 27790387 DOI: 10.1080/21659087.2016.1187803] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor hypoxia is linked to tumor progression, metastasis, and therapy resistance. However, the underlying mechanisms behind this linkage are not fully understood. Here we present a novel fluorescent mCherry hypoxia-responsive marker that can be used in real time imaging to specifically and sensitively identify hypoxic cells in vivo at single cell resolution. Tumors derived from triple negative tumor cells expressing the hypoxia marker reveal that the hypoxic tumor cells congregate near flowing blood vessels. Using multiphoton microscopy, hypoxic MDA-MB-231 cells were directly visualized and showed a more persistent slow migration phenotype as compared to normoxic cells in the same field in vivo. Hypoxic tumor cells are enriched in the cell population that migrates toward human epithelial growth factor gradients in vivo, and has increased collagen degradation and intravasation activity, characteristics of dissemination and metastasis competent tumor cells. The hypoxia probe introduced in this study provides a specific reporter of hypoxic cell phenotypes in vivo which reveals new insights into the mechanisms by which hypoxia is linked to metastasis.
Collapse
Affiliation(s)
- Yarong Wang
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA; Integrated Imaging Program; Albert Einstein College of Medicine,Bronx, New York, USA
| | - Haoxuan Wang
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA
| | - Jiufeng Li
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - David Entenberg
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA; Gruss Lipper Biophotonics Center; Albert Einstein College of Medicine; Bronx, NY USA; Integrated Imaging Program; Albert Einstein College of Medicine,Bronx, New York, USA
| | - Alice Xue
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA
| | - Weigang Wang
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA
| | - John Condeelis
- Department of Anatomy and Structural Biology; Albert Einstein College of Medicine; Bronx, NY USA; Gruss Lipper Biophotonics Center; Albert Einstein College of Medicine; Bronx, NY USA; Integrated Imaging Program; Albert Einstein College of Medicine,Bronx, New York, USA
| |
Collapse
|
87
|
Abstract
Metastasis is the underlying cause of death for the majority of breast cancer patients. Despite significant advances in recent years in basic research and clinical development, therapies that specifically target metastatic breast cancer remain inadequate, and represents the single greatest obstacle to reducing mortality of late-stage breast cancer. Recent efforts have leveraged genomic analysis of breast cancer and molecular dissection of tumor-stromal cross-talk to uncover a number of promising candidates for targeted treatment of metastatic breast cancer. Rational combinations of therapeutic agents targeting tumor-intrinsic properties and microenvironmental components provide a promising strategy to develop precision treatments with higher specificity and less toxicity. In this review, we discuss the emerging therapeutic targets in breast cancer metastasis, from tumor-intrinsic pathways to those that involve the host tissue components, including the immune system.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, United States.
| |
Collapse
|
88
|
Parsons B, Foley E. Cellular immune defenses of Drosophila melanogaster. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 58:95-101. [PMID: 26748247 DOI: 10.1016/j.dci.2015.12.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/23/2015] [Accepted: 12/23/2015] [Indexed: 06/05/2023]
Abstract
Drosophila melanogaster is a widely used model for the characterization of blood cell development and function, with an array of protocols for the manipulation and visualization of fixed or live cells in vitro or in vivo. Researchers have deployed these techniques to reveal Drosophila hemocytes as a remarkably versatile cell type that engulfs apoptotic corpses; neutralizes invading parasites; seals epithelial wounds; and deposits extracellular matrix proteins. In this review, we will discuss the key features of Drosophila hemocyte development and function, and identify similarities with vertebrate counterparts.
Collapse
Affiliation(s)
- Brendon Parsons
- 1B3.14, 8440-112 Street, Walter Mackenzie Health Sciences Centre, University of Alberta, Edmonton, AB, T6G 2J2, Canada
| | - Edan Foley
- University of Alberta, Department of Medical Microbiology and Immunology, Canada.
| |
Collapse
|
89
|
Karagiannis GS, Goswami S, Jones JG, Oktay MH, Condeelis JS. Signatures of breast cancer metastasis at a glance. J Cell Sci 2016; 129:1751-8. [PMID: 27084578 DOI: 10.1242/jcs.183129] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gene expression profiling has yielded expression signatures from which prognostic tests can be derived to facilitate clinical decision making in breast cancer patients. Some of these signatures are based on profiling of whole tumor tissue (tissue signatures), which includes all tumor and stromal cells. Prognostic markers have also been derived from the profiling of metastasizing tumor cells, including circulating tumor cells (CTCs) and migratory-disseminating tumor cells within the primary tumor. The metastasis signatures based on CTCs and migratory-disseminating tumor cells have greater potential for unraveling cell biology insights and mechanistic underpinnings of tumor cell dissemination and metastasis. Of clinical interest is the promise that stratification of patients into high or low metastatic risk, as well as assessing the need for cytotoxic therapy, might be improved if prognostics derived from these two types of signatures are used in a combined way. The aim of this Cell Science at a Glance article and accompanying poster is to navigate through both types of signatures and their derived prognostics, as well as to highlight biological insights and clinical applications that could be derived from them, especially when they are used in combination.
Collapse
Affiliation(s)
- George S Karagiannis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sumanta Goswami
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Joan G Jones
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Maja H Oktay
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY 10461, USA Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
90
|
Abstract
The process of entering the bloodstream, intravasation, is a necessary step in the development of distant metastases. The focus of this review is on the pathways and molecules that have been identified as being important based on current in vitro and in vivo assays for intravasation. Properties of the vasculature which are important for intravasation include microvessel density and also diameter of the vasculature, with increased intravasation correlating with increased vessel diameter in some tumors. TGFB signaling can enhance intravasation at least in part through induction of EMT, and we discuss other TGFB target genes that are important for intravasation. In addition to TGFB signaling, a number of studies have demonstrated that activation of EGF receptor family members stimulates intravasation, with downstream signaling through PI3K, N-WASP, RhoA, and WASP to induce invadopodia. With respect to proteases, there is strong evidence for contributions by uPA/uPAR, while the roles of MMPs in intravasation may be more tumor specific. Other cells including macrophages, fibroblasts, neutrophils, and platelets can also play a role in enhancing tumor cell intravasation. The technology is now available to interrogate the expression patterns of circulating tumor cells, which will provide an important reality check for the model systems being used. With a better understanding of the mechanisms underlying intravasation, the goal is to provide new opportunities for improving prognosis as well as potentially developing new treatments.
Collapse
Affiliation(s)
- Serena P H Chiang
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Ramon M Cabrera
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Jeffrey E Segall
- Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
91
|
Di Martino J, Henriet E, Ezzoukhry Z, Goetz JG, Moreau V, Saltel F. The microenvironment controls invadosome plasticity. J Cell Sci 2016; 129:1759-68. [PMID: 27029343 DOI: 10.1242/jcs.182329] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Invadosomes are actin-based structures involved in extracellular matrix degradation. Invadosomes is a term that includes podosomes and invadopodia, which decorate normal and tumour cells, respectively. They are mainly organised into dots or rosettes, and podosomes and invadopodia are often compared and contrasted. Various internal or external stimuli have been shown to induce their formation and/or activity. In this Commentary, we address the impact of the microenvironment and the role of matrix receptors on the formation, and dynamic and degradative activities of invadosomes. In particular, we highlight recent findings regarding the role of type I collagen fibrils in inducing the formation of a new linear organisation of invadosomes. We will also discuss invadosome plasticity more generally and emphasise its physio-pathological relevance.
Collapse
Affiliation(s)
- Julie Di Martino
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| | - Elodie Henriet
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| | - Zakaria Ezzoukhry
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| | - Jacky G Goetz
- MN3T, Inserm U1109, Strasbourg 67200, France Université de Strasbourg, Strasbourg 67000, France LabEx Medalis, Université de Strasbourg, Strasbourg 67000, France Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg 67000, France
| | - Violaine Moreau
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| | - Frederic Saltel
- Institut National de la Santé et de la Recherche Médicale, U1053, Bordeaux F-33076, France Université de Bordeaux, Bordeaux F-33076, France
| |
Collapse
|
92
|
Scheele CLGJ, Maynard C, van Rheenen J. Intravital Insights into Heterogeneity, Metastasis, and Therapy Responses. Trends Cancer 2016; 2:205-216. [PMID: 28741572 DOI: 10.1016/j.trecan.2016.03.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 01/08/2023]
Abstract
Tumor progression is driven by a series of genetic and microenvironmental changes. These events lead to heterogeneous tumors which consist of a variety of cells from which some cells may possess properties which promote survival after therapy and metastasis. Recent advances in intravital microscopy (IVM) have enabled visualization of this tumor heterogeneity over time at a single-cell resolution. We highlight here the latest IVM studies that have revealed the dynamic interactions between the tumor cells and their local microenvironment. We review the most recent data that exposes how these dynamic interactions cause an additional increase in tumor heterogeneity, resulting in multiple metastatic strategies and facilitating therapy resistance.
Collapse
Affiliation(s)
- Colinda L G J Scheele
- Cancer Genomics Netherlands, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), and University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Carrie Maynard
- Cancer Genomics Netherlands, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), and University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jacco van Rheenen
- Cancer Genomics Netherlands, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), and University Medical Centre Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
93
|
Carmona G, Perera U, Gillett C, Naba A, Law AL, Sharma VP, Wang J, Wyckoff J, Balsamo M, Mosis F, De Piano M, Monypenny J, Woodman N, McConnell RE, Mouneimne G, Van Hemelrijck M, Cao Y, Condeelis J, Hynes RO, Gertler FB, Krause M. Lamellipodin promotes invasive 3D cancer cell migration via regulated interactions with Ena/VASP and SCAR/WAVE. Oncogene 2016; 35:5155-69. [PMID: 26996666 PMCID: PMC5031503 DOI: 10.1038/onc.2016.47] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 01/20/2016] [Accepted: 02/08/2016] [Indexed: 12/16/2022]
Abstract
Cancer invasion is a hallmark of metastasis. The mesenchymal mode of cancer cell invasion is mediated by elongated membrane protrusions driven by the assembly of branched F-actin networks. How deregulation of actin regulators promotes cancer cell invasion is still enigmatic. We report that increased expression and membrane localization of the actin regulator Lamellipodin correlate with reduced metastasis-free survival and poor prognosis in breast cancer patients. In agreement, we find that Lamellipodin depletion reduced lung metastasis in an orthotopic mouse breast cancer model. Invasive 3D cancer cell migration as well as invadopodia formation and matrix degradation was impaired upon Lamellipodin depletion. Mechanistically, we show that Lamellipodin promotes invasive 3D cancer cell migration via both actin-elongating Ena/VASP proteins and the Scar/WAVE complex, which stimulates actin branching. In contrast, Lamellipodin interaction with Scar/WAVE but not with Ena/VASP is required for random 2D cell migration. We identified a phosphorylation-dependent mechanism that regulates selective recruitment of these effectors to Lamellipodin: Abl-mediated Lamellipodin phosphorylation promotes its association with both Scar/WAVE and Ena/VASP, whereas Src-dependent phosphorylation enhances binding to Scar/WAVE but not to Ena/VASP. Through these selective, regulated interactions Lamellipodin mediates directional sensing of epidermal growth factor (EGF) gradients and invasive 3D migration of breast cancer cells. Our findings imply that increased Lamellipodin levels enhance Ena/VASP and Scar/WAVE activities at the plasma membrane to promote 3D invasion and metastasis.
Collapse
Affiliation(s)
- G Carmona
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - U Perera
- King's College London, Randall Division of Cell and Molecular Biophysics, London, UK
| | - C Gillett
- King's College London, Research Oncology, Division of Cancer Studies, Faculty of Life Sciences and Medicine, London, UK
| | - A Naba
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - A-L Law
- King's College London, Randall Division of Cell and Molecular Biophysics, London, UK
| | - V P Sharma
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA.,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Wang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - J Wyckoff
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - M Balsamo
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - F Mosis
- King's College London, Randall Division of Cell and Molecular Biophysics, London, UK
| | - M De Piano
- King's College London, Division of Cancer Studies, Cancer Epidemiology Group, London, UK
| | - J Monypenny
- King's College London, Randall Division of Cell and Molecular Biophysics, London, UK.,King's College London, Research Oncology, Division of Cancer Studies, Faculty of Life Sciences and Medicine, London, UK.,King's College London, Division of Cancer Studies, Richard Dimbleby Department of Cancer Research, London, UK
| | - N Woodman
- King's College London, Research Oncology, Division of Cancer Studies, Faculty of Life Sciences and Medicine, London, UK
| | - R E McConnell
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - G Mouneimne
- University of Arizona Cancer Center, Tucson, AZ, USA
| | - M Van Hemelrijck
- King's College London, Division of Cancer Studies, Cancer Epidemiology Group, London, UK
| | - Y Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - J Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, USA.,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - R O Hynes
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - F B Gertler
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - M Krause
- King's College London, Randall Division of Cell and Molecular Biophysics, London, UK
| |
Collapse
|
94
|
Beerling E, Seinstra D, de Wit E, Kester L, van der Velden D, Maynard C, Schäfer R, van Diest P, Voest E, van Oudenaarden A, Vrisekoop N, van Rheenen J. Plasticity between Epithelial and Mesenchymal States Unlinks EMT from Metastasis-Enhancing Stem Cell Capacity. Cell Rep 2016; 14:2281-8. [PMID: 26947068 PMCID: PMC4802222 DOI: 10.1016/j.celrep.2016.02.034] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/16/2015] [Accepted: 02/02/2016] [Indexed: 12/20/2022] Open
Abstract
Forced overexpression and/or downregulation of proteins regulating epithelial-to-mesenchymal transition (EMT) has been reported to alter metastasis by changing migration and stem cell capacity of tumor cells. However, these manipulations artificially keep cells in fixed states, while in vivo cells may adapt transient and reversible states. Here, we have tested the existence and role of epithelial-mesenchymal plasticity in metastasis of mammary tumors without artificially modifying EMT regulators. In these tumors, we found by intravital microscopy that the motile tumor cells have undergone EMT, while their epithelial counterparts were not migratory. Moreover, we found that epithelial-mesenchymal plasticity renders any EMT-induced stemness differences, as reported previously, irrelevant for metastatic outgrowth, because mesenchymal cells that arrive at secondary sites convert to the epithelial state within one or two divisions, thereby obtaining the same stem cell potential as their arrived epithelial counterparts. We conclude that epithelial-mesenchymal plasticity supports migration but additionally eliminates stemness-enhanced metastatic outgrowth differences. Direct evidence of EMT obtained in unperturbed breast tumors by real-time visualization EMT exists in breast tumors without experimentally altering EMT inducers Tumor cells that underwent EMT are the migratory cells within a tumor Outgrowth potential differences between states are irrelevant due to plasticity
Collapse
Affiliation(s)
- Evelyne Beerling
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Daniëlle Seinstra
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Elzo de Wit
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands; Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Lennart Kester
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | | | - Carrie Maynard
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Ronny Schäfer
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Paul van Diest
- Department of Pathology, University Medical Centre Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands
| | - Emile Voest
- Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | - Alexander van Oudenaarden
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Nienke Vrisekoop
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands.
| | - Jacco van Rheenen
- Cancer Genomics Center-Hubrecht Institute-KNAW & University Medical Centre Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands.
| |
Collapse
|
95
|
Oudin MJ, Jonas O, Kosciuk T, Broye LC, Guido BC, Wyckoff J, Riquelme D, Lamar JM, Asokan SB, Whittaker C, Ma D, Langer R, Cima MJ, Wisinski KB, Hynes RO, Lauffenburger DA, Keely PJ, Bear JE, Gertler FB. Tumor Cell-Driven Extracellular Matrix Remodeling Drives Haptotaxis during Metastatic Progression. Cancer Discov 2016; 6:516-31. [PMID: 26811325 DOI: 10.1158/2159-8290.cd-15-1183] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/21/2016] [Indexed: 12/14/2022]
Abstract
UNLABELLED Fibronectin (FN) is a major component of the tumor microenvironment, but its role in promoting metastasis is incompletely understood. Here, we show that FN gradients elicit directional movement of breast cancer cells, in vitro and in vivo Haptotaxis on FN gradients requires direct interaction between α5β1 integrin and MENA, an actin regulator, and involves increases in focal complex signaling and tumor cell-mediated extracellular matrix (ECM) remodeling. Compared with MENA, higher levels of the prometastatic MENA(INV) isoform associate with α5, which enables 3-D haptotaxis of tumor cells toward the high FN concentrations typically present in perivascular space and in the periphery of breast tumor tissue. MENA(INV) and FN levels were correlated in two breast cancer cohorts, and high levels of MENA(INV) were significantly associated with increased tumor recurrence as well as decreased patient survival. Our results identify a novel tumor cell-intrinsic mechanism that promotes metastasis through ECM remodeling and ECM-guided directional migration. SIGNIFICANCE Here, we provide new insight into how tumor cell:ECM interactions generate signals and structures that promote directed tumor cell migration, a critical component of metastasis. Our results identify a tumor cell-intrinsic mechanism driven by the actin regulatory protein MENA that promotes ECM remodeling and haptotaxis along FN gradients. Cancer Discov; 6(5); 516-31. ©2016 AACR.See related commentary by Santiago-Medina and Yang, p. 474This article is highlighted in the In This Issue feature, p. 461.
Collapse
Affiliation(s)
- Madeleine J Oudin
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Oliver Jonas
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Tatsiana Kosciuk
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Liliane C Broye
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Bruna C Guido
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Jeff Wyckoff
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Daisy Riquelme
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - John M Lamar
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Sreeja B Asokan
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, North Carolina
| | - Charlie Whittaker
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Duanduan Ma
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Michael J Cima
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts
| | - Kari B Wisinski
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, Wisconsin
| | - Richard O Hynes
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts. Department of Biology, MIT, Cambridge, Massachusetts. Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Douglas A Lauffenburger
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts. Department of Biological Engineering, MIT, Cambridge, Massachusetts
| | - Patricia J Keely
- Department of Cell and Regenerative Biology, University of Wisconsin Madison, Madison, Wisconsin
| | - James E Bear
- Lineberger Comprehensive Cancer Center, UNC Chapel Hill, Chapel Hill, North Carolina. Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Frank B Gertler
- David H. Koch Institute for Integrative Cancer Research, MIT, Cambridge, Massachusetts. Department of Biology, MIT, Cambridge, Massachusetts.
| |
Collapse
|
96
|
Knutsdottir H, Condeelis JS, Palsson E. 3-D individual cell based computational modeling of tumor cell-macrophage paracrine signaling mediated by EGF and CSF-1 gradients. Integr Biol (Camb) 2015; 8:104-19. [PMID: 26686751 DOI: 10.1039/c5ib00201j] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
High density of macrophages in mammary tumors has been associated with a higher risk of metastasis and thus increased mortality in women. The EGF/CSF-1 paracrine signaling increases the number of invasive tumor cells by both recruiting tumor cells further away and manipulating the macrophages' innate ability to open up a passage into blood vessels thus promoting intravasation and finally metastasis. A 3-D individual-cell-based model is introduced, to better understand the tumor cell-macrophage interactions, and to explore how changing parameters of the paracrine signaling system affects the number of invasive tumor cells. The simulation data and videos of the cell movements correlated well with findings from both in vitro and in vivo experimental results. The model demonstrated how paracrine signaling is necessary to achieve co-migration of tumor cells and macrophages towards a specific signaling source. We showed how the paracrine signaling enhances the number of both invasive tumor cells and macrophages. The simulations revealed that for the in vitro experiments the imposed no-flux boundary condition might be affecting the results, and that changing the setup might lead to different experimental findings. In our simulations, the 3 : 1 tumor cell/macrophage ratio, observed in vivo, was robust for many parameters but sensitive to EGF signal strength and fraction of macrophages in the tumor. The model can be used to identify new agents for targeted therapy and we suggest that a successful strategy to prevent or limit invasion of tumor cells would be to block the tumor cell-macrophage paracrine signaling. This can be achieved by either blocking the EGF or CSF-1 receptors or supressing the EGF or CSF-1 signal.
Collapse
Affiliation(s)
- Hildur Knutsdottir
- Mathematics Department/Institute of Applied Mathematics, University of British Columbia, Vancouver, BC V6 T 1Z2, Canada
| | | | | |
Collapse
|
97
|
Oudin MJ, Hughes SK, Rohani N, Moufarrej MN, Jones JG, Condeelis JS, Lauffenburger DA, Gertler FB. Characterization of the expression of the pro-metastatic Mena(INV) isoform during breast tumor progression. Clin Exp Metastasis 2015; 33:249-61. [PMID: 26680363 DOI: 10.1007/s10585-015-9775-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/07/2015] [Indexed: 01/16/2023]
Abstract
Several functionally distinct isoforms of the actin regulatory Mena are produced by alternative splicing during tumor progression. Forced expression of the Mena(INV) isoform drives invasion, intravasation and metastasis. However, the abundance and distribution of endogenously expressed Mena(INV) within primary tumors during progression remain unknown, as most studies to date have only assessed relative mRNA levels from dissociated tumor samples. We have developed a Mena(INV) isoform-specific monoclonal antibody and used it to examine Mena(INV) expression patterns in mouse mammary and human breast tumors. Mena(INV) expression increases during tumor progression and to examine the relationship between Mena(INV) expression and markers for epithelial or mesenchymal status, stemness, stromal cell types and hypoxic regions. Further, while Mena(INV) robustly expressed in vascularized areas of the tumor, it is not confined to cells adjacent to blood vessels. Altogether, these data demonstrate the specificity and utility of the anti-Mena(INV)-isoform specific antibody, and provide the first description of endogenous Mena(INV) protein expression in mouse and human tumors.
Collapse
Affiliation(s)
- Madeleine J Oudin
- Koch Institute for Integrative Cancer Research, MIT, 76-317, 77 Massachusetts Ave, Cambridge, MA, 02139, USA.
| | - Shannon K Hughes
- Koch Institute for Integrative Cancer Research, MIT, 76-317, 77 Massachusetts Ave, Cambridge, MA, 02139, USA.,Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA
| | - Nazanin Rohani
- Koch Institute for Integrative Cancer Research, MIT, 76-317, 77 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Mira N Moufarrej
- Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA
| | - Joan G Jones
- Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Douglas A Lauffenburger
- Koch Institute for Integrative Cancer Research, MIT, 76-317, 77 Massachusetts Ave, Cambridge, MA, 02139, USA.,Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA
| | - Frank B Gertler
- Koch Institute for Integrative Cancer Research, MIT, 76-317, 77 Massachusetts Ave, Cambridge, MA, 02139, USA.,Department of Biology, MIT, Cambridge, MA, 02139, USA
| |
Collapse
|
98
|
Choi M, Kwok SJJ, Yun SH. In vivo fluorescence microscopy: lessons from observing cell behavior in their native environment. Physiology (Bethesda) 2015; 30:40-9. [PMID: 25559154 DOI: 10.1152/physiol.00019.2014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microscopic imaging techniques to visualize cellular behaviors in their natural environment play a pivotal role in biomedical research. Here, we review how recent technical advances in intravital microscopy have enabled unprecedented access to cellular physiology in various organs of mice in normal and diseased states.
Collapse
Affiliation(s)
- Myunghwan Choi
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts; and
| | - Sheldon J J Kwok
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts; and Harvard-MIT Health Sciences and Technology, Cambridge, Massachusetts
| | - Seok Hyun Yun
- Harvard Medical School and Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts; and Harvard-MIT Health Sciences and Technology, Cambridge, Massachusetts
| |
Collapse
|
99
|
Hughes SK, Oudin MJ, Tadros J, Neil J, Del Rosario A, Joughin BA, Ritsma L, Wyckoff J, Vasile E, Eddy R, Philippar U, Lussiez A, Condeelis JS, van Rheenen J, White F, Lauffenburger DA, Gertler FB. PTP1B-dependent regulation of receptor tyrosine kinase signaling by the actin-binding protein Mena. Mol Biol Cell 2015; 26:3867-78. [PMID: 26337385 PMCID: PMC4626070 DOI: 10.1091/mbc.e15-06-0442] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/25/2015] [Indexed: 12/17/2022] Open
Abstract
The actin-binding protein Mena regulates RTK signaling after growth factor stimulation in tumor cells by a novel mechanism. The alternatively spliced MenaINV isoform disrupts this attenuation to drive sensitivity to growth factors, resistance to targeted inhibitors, and ultimately tumor invasion and metastasis. During breast cancer progression, alternative mRNA splicing produces functionally distinct isoforms of Mena, an actin regulator with roles in cell migration and metastasis. Aggressive tumor cell subpopulations express MenaINV, which promotes tumor cell invasion by potentiating EGF responses. However, the mechanism by which this occurs is unknown. Here we report that Mena associates constitutively with the tyrosine phosphatase PTP1B and mediates a novel negative feedback mechanism that attenuates receptor tyrosine kinase signaling. On EGF stimulation, complexes containing Mena and PTP1B are recruited to the EGFR, causing receptor dephosphorylation and leading to decreased motility responses. Mena also interacts with the 5′ inositol phosphatase SHIP2, which is important for the recruitment of the Mena-PTP1B complex to the EGFR. When MenaINV is expressed, PTP1B recruitment to the EGFR is impaired, providing a mechanism for growth factor sensitization to EGF, as well as HGF and IGF, and increased resistance to EGFR and Met inhibitors in signaling and motility assays. In sum, we demonstrate that Mena plays an important role in regulating growth factor–induced signaling. Disruption of this attenuation by MenaINV sensitizes tumor cells to low–growth factor concentrations, thereby increasing the migration and invasion responses that contribute to aggressive, malignant cell phenotypes.
Collapse
Affiliation(s)
- Shannon K Hughes
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Madeleine J Oudin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jenny Tadros
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jason Neil
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Amanda Del Rosario
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brian A Joughin
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Laila Ritsma
- Cancer Genomics Netherlands-Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584 CX Utrecht, Netherlands
| | - Jeff Wyckoff
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, NY 10461
| | - Eliza Vasile
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Robert Eddy
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, NY 10461
| | - Ulrike Philippar
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Alisha Lussiez
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, New York, NY 10461
| | - Jacco van Rheenen
- Cancer Genomics Netherlands-Hubrecht Institute-KNAW and University Medical Centre Utrecht, 3584 CX Utrecht, Netherlands
| | - Forest White
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Douglas A Lauffenburger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Frank B Gertler
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
100
|
Rua R, McGavern DB. Elucidation of monocyte/macrophage dynamics and function by intravital imaging. J Leukoc Biol 2015; 98:319-32. [PMID: 26162402 PMCID: PMC4763596 DOI: 10.1189/jlb.4ri0115-006rr] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 06/19/2015] [Accepted: 06/23/2015] [Indexed: 12/21/2022] Open
Abstract
Monocytes and macrophages are a diverse population of innate immune cells that play a critical role in homeostasis and inflammation. These cells are surveillant by nature and closely monitor the vasculature and surrounding tissue during states of health and disease. Given their abundance and strategic positioning throughout the body, myeloid cells are among the first responders to any inflammatory challenge and are active participants in most immune-mediated diseases. Recent studies have shed new light on myeloid cell dynamics and function by use of an imaging technique referred to as intravital microscopy (IVM). This powerful approach allows researchers to gain real-time insights into monocytes and macrophages performing homeostatic and inflammatory tasks in living tissues. In this review, we will present a contemporary synopsis of how intravital microscopy has revolutionized our understanding of myeloid cell contributions to vascular maintenance, microbial defense, autoimmunity, tumorigenesis, and acute/chronic inflammatory diseases.
Collapse
Affiliation(s)
- Rejane Rua
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|