51
|
Follain G, Gensbittel V, Mary B, Lefebvre O, Harlepp S, Hyenne V, Goetz JG. [Influence of fluid mechanics on metastasis formation]. Med Sci (Paris) 2020; 36:872-878. [PMID: 33026329 DOI: 10.1051/medsci/2020158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Metastases are the main cause of cancer-related deaths. The chain of events leading to their development is called "the metastatic cascade". The biological and biochemical aspects of this process have been well studied but the importance of biomechanical parameters only recently became a focus in the field. Studies have shown the biological fluids (blood, lymph and interstitial fluid) to play a key role in the metastatic cascade. These fluids participate in the transport of circulating tumor cells (CTCs) as well as the factors that they secrete, while at the same time influencing the events of the metastatic cascade through the forces that they generate. The hemodynamic properties and topological constraints of the vascular architecture control the formation of metastatic niches and the metastatic potential of tumor cells. In this review, we discuss the importance of these mechanical forces and highlight the novel questions and research avenues that they open.
Collapse
Affiliation(s)
- Gautier Follain
- Inserm UMR_S1109, 1 place de l'Hôpital, F-67000 Strasbourg, France - Université de Strasbourg, F-67000 Strasbourg, France - Fédération de médecine translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France - Adresse actuelle : Turku Bioscience Center, University of Turku and Åbo Akademi University, FI-20520, Turku, Finlande
| | - Valentin Gensbittel
- Inserm UMR_S1109, 1 place de l'Hôpital, F-67000 Strasbourg, France - Université de Strasbourg, F-67000 Strasbourg, France - Fédération de médecine translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Benjamin Mary
- Inserm UMR_S1109, 1 place de l'Hôpital, F-67000 Strasbourg, France - Université de Strasbourg, F-67000 Strasbourg, France - Fédération de médecine translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Olivier Lefebvre
- Inserm UMR_S1109, 1 place de l'Hôpital, F-67000 Strasbourg, France - Université de Strasbourg, F-67000 Strasbourg, France - Fédération de médecine translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Sébastien Harlepp
- Inserm UMR_S1109, 1 place de l'Hôpital, F-67000 Strasbourg, France - Université de Strasbourg, F-67000 Strasbourg, France - Fédération de médecine translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| | - Vincent Hyenne
- Inserm UMR_S1109, 1 place de l'Hôpital, F-67000 Strasbourg, France - Université de Strasbourg, F-67000 Strasbourg, France - Fédération de médecine translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France - CNRS, SNC 5055,
| | - Jacky G Goetz
- Inserm UMR_S1109, 1 place de l'Hôpital, F-67000 Strasbourg, France - Université de Strasbourg, F-67000 Strasbourg, France - Fédération de médecine translationnelle de Strasbourg (FMTS), F-67000 Strasbourg, France
| |
Collapse
|
52
|
Izraely S, Witz IP. Site-specific metastasis: A cooperation between cancer cells and the metastatic microenvironment. Int J Cancer 2020; 148:1308-1322. [PMID: 32761606 PMCID: PMC7891572 DOI: 10.1002/ijc.33247] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 07/08/2020] [Accepted: 08/03/2020] [Indexed: 12/19/2022]
Abstract
The conclusion derived from the information provided in this review is that disseminating tumor cells (DTC) collaborate with the microenvironment of a future metastatic organ site in the establishment of organ‐specific metastasis. We review the basic principles of site‐specific metastasis and the contribution of the cross talk between DTC and the microenvironment of metastatic sites (metastatic microenvironment [MME]) to the establishment of the organ‐specific premetastatic niche; the targeted migration of DTC to the endothelium of the future organ‐specific metastasis; the transmigration of DTC to this site and the seeding and colonization of DTC in their future MME. We also discuss the role played by DTC‐MME interactions on tumor dormancy and on the differential response of tumor cells residing in different MMEs to antitumor therapy. Finally, we summarize some studies dealing with the effects of the MME on a unique site‐specific metastasis—brain metastasis.
Collapse
Affiliation(s)
- Sivan Izraely
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Isaac P Witz
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| |
Collapse
|
53
|
Aguirre LA, Montalbán-Hernández K, Avendaño-Ortiz J, Marín E, Lozano R, Toledano V, Sánchez-Maroto L, Terrón V, Valentín J, Pulido E, Casalvilla JC, Rubio C, Diekhorst L, Laso-García F, del Fresno C, Collazo-Lorduy A, Jiménez-Munarriz B, Gómez-Campelo P, Llanos-González E, Fernández-Velasco M, Rodríguez-Antolín C, Pérez de Diego R, Cantero-Cid R, Hernádez-Jimenez E, Álvarez E, Rosas R, dies López-Ayllón B, de Castro J, Wculek SK, Cubillos-Zapata C, Ibáñez de Cáceres I, Díaz-Agero P, Gutiérrez Fernández M, Paz de Miguel M, Sancho D, Schulte L, Perona R, Belda-Iniesta C, Boscá L, López-Collazo E. Tumor stem cells fuse with monocytes to form highly invasive tumor-hybrid cells. Oncoimmunology 2020; 9:1773204. [PMID: 32923132 PMCID: PMC7458638 DOI: 10.1080/2162402x.2020.1773204] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/14/2020] [Accepted: 05/10/2020] [Indexed: 02/07/2023] Open
Abstract
The 'cancer cell fusion' theory is controversial due to the lack of methods available to identify hybrid cells and to follow the phenomenon in patients. However, it seems to be one of the best explanations for both the origin and metastasis of primary tumors. Herein, we co-cultured lung cancer stem cells with human monocytes and analyzed the dynamics and properties of tumor-hybrid cells (THC), as well as the molecular mechanisms beneath this fusion process by several techniques: electron-microscopy, karyotyping, CRISPR-Cas9, RNA-seq, immunostaining, signaling blockage, among others. Moreover, mice models were assessed for in vivo characterization of hybrids colonization and invasiveness. Then, the presence of THCs in bloodstream and samples from primary and metastatic lesions were detected by FACS and immunofluorescence protocols, and their correlations with TNM stages established. Our data indicate that the generation of THCs depends on the expression of CD36 on tumor stem cells and the oxidative state and polarization of monocytes, the latter being strongly influenced by microenvironmental fluctuations. Highly oxidized M2-like monocytes show the strongest affinity to fuse with tumor stem cells. THCs are able to proliferate, colonize and invade organs. THC-specific cell surface signature CD36+CD14+PANK+ allows identifying them in matched primary tumor tissues and metastases as well as in bloodstream from patients with lung cancer, thus functioning as a biomarker. THCs levels in circulation correlate with TNM classification. Our results suggest that THCs are involved in both origin and spread of metastatic cells. Furthermore, they might set the bases for future therapies to avoid or eradicate lung cancer metastasis.
Collapse
Affiliation(s)
- Luis Augusto Aguirre
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Karla Montalbán-Hernández
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - José Avendaño-Ortiz
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Centre for Biomedical Research Network of Respiratory Diseases (CIBERES), Madrid, Spain
| | - Elvira Marín
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Roberto Lozano
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Víctor Toledano
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Centre for Biomedical Research Network of Respiratory Diseases (CIBERES), Madrid, Spain
| | - Laura Sánchez-Maroto
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Verónica Terrón
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Jaime Valentín
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Elisa Pulido
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - José Carlos Casalvilla
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Carolina Rubio
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Luke Diekhorst
- Department of Neurology and Stroke Centre, Neuroscience and Cerebrovascular Research Laboratory, IdiPAZ, La Paz University Hospital, Autonomous University of Madrid, Madrid, Spain
| | - Fernando Laso-García
- Department of Neurology and Stroke Centre, Neuroscience and Cerebrovascular Research Laboratory, IdiPAZ, La Paz University Hospital, Autonomous University of Madrid, Madrid, Spain
| | - Carlos del Fresno
- Immunobiology Laboratory, National Centre for Cardiovascular Research (CNIC), Madrid, Spain
| | | | | | - Paloma Gómez-Campelo
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Emilio Llanos-González
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - María Fernández-Velasco
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Centre for Biomedical Research Network, CIBER-CV, Madrid, Spain
| | - Carlos Rodríguez-Antolín
- Biomarkers and Experimental Therapeutics in Cancer Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Rebeca Pérez de Diego
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Laboratory of Immunogenetics of Human Diseases, IdiPAZ, Madrid, Spain
| | - Ramón Cantero-Cid
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Enrique Hernádez-Jimenez
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Enrique Álvarez
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Rocío Rosas
- Biomarkers and Experimental Therapeutics in Cancer Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Blanca dies López-Ayllón
- Biomarkers and Experimental Therapeutics in Cancer Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Laboratory of Experimental Models of Human Diseases, Biomedical Research Institute CSIC/UAM, Madrid, Spain
- Centre for Biomedical Research Network, CIBERER, Madrid, Spain
| | - Javier de Castro
- Biomarkers and Experimental Therapeutics in Cancer Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - Stefanie K. Wculek
- Immunobiology Laboratory, National Centre for Cardiovascular Research (CNIC), Madrid, Spain
| | - Carolina Cubillos-Zapata
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Centre for Biomedical Research Network of Respiratory Diseases (CIBERES), Madrid, Spain
| | | | | | - María Gutiérrez Fernández
- Department of Neurology and Stroke Centre, Neuroscience and Cerebrovascular Research Laboratory, IdiPAZ, La Paz University Hospital, Autonomous University of Madrid, Madrid, Spain
| | - María Paz de Miguel
- Cell Engineering Laboratory, IdiPAZ, La Paz University Hospital, Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, National Centre for Cardiovascular Research (CNIC), Madrid, Spain
| | - Leon Schulte
- Institute for Lung Research/iLung, Research Group “Rna-biology of Inflammation & Infection,” Philipps University, Marburg, Germany
| | - Rosario Perona
- Biomarkers and Experimental Therapeutics in Cancer Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Laboratory of Experimental Models of Human Diseases, Biomedical Research Institute CSIC/UAM, Madrid, Spain
- Centre for Biomedical Research Network, CIBERER, Madrid, Spain
| | | | - Lisardo Boscá
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Centre for Biomedical Research Network, CIBER-CV, Madrid, Spain
- Laboratory of Experimental Models of Human Diseases, Biomedical Research Institute CSIC/UAM, Madrid, Spain
| | - Eduardo López-Collazo
- The Innate Immune Response Group, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Tumour Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain
- Centre for Biomedical Research Network of Respiratory Diseases (CIBERES), Madrid, Spain
| |
Collapse
|
54
|
Abstract
Cell migration is highly sensitive to fluid shear stress (FSS) in blood flow or interstitial fluid flow. However, whether the FSS gradient can regulate the migration of cells remains unclear. In this work, we constructed a parallel-plate flow chamber with different FSS gradients and verified the gradient flow field by particle image velocimetry measurements and finite element analyses. We then investigated the effect of FSS magnitudes and gradients on the migration of osteoclast precursor RAW264.7 cells. Results showed that the cells sensed the FSS gradient and migrated toward the low-FSS region. This FSS gradient-induced migration tended to occur in low-FSS magnitudes and high gradients, e.g., the migration angle relative to flow direction was approximately 90° for 0.1 Pa FSS and 0.2 Pa mm−1 FSS gradient. When chemically inhibiting the calcium signaling pathways of the mechanosensitive cation channel, endoplasmic reticulum, phospholipase C, and extracellular calcium, the cell migration toward the low-FSS region was significantly reduced. This study may provide insights into the mechanism of the recruitment of osteoclast precursors at the site of bone resorption and of mechanical stimulation-induced bone remodeling.
Collapse
Affiliation(s)
- Yan Gao
- a Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering , Beijing Institute of Technology , Beijing , P. R. China
| | - Taiyang Li
- a Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering , Beijing Institute of Technology , Beijing , P. R. China
| | - Qing Sun
- a Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering , Beijing Institute of Technology , Beijing , P. R. China
| | - Bo Huo
- a Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering , Beijing Institute of Technology , Beijing , P. R. China
| |
Collapse
|
55
|
Rajabi S, Shakib H, Dastmalchi R, Danesh-Afrooz A, Karima S, Hedayati M. Metastatic propagation of thyroid cancer; organ tropism and major modulators. Future Oncol 2020; 16:1301-1319. [PMID: 32421354 DOI: 10.2217/fon-2019-0780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Thyroid cancer, as the most prevalent endocrine malignancy, comprises nearly 1% of all cancers in the world. The metastatic propagation of thyroid cancer is under the control of a number of modulating processes and factors such as signaling pathways and their components, cell division regulators, metabolic reprogramming factors, extracellular matrix remodelers, epithelial to mesenchymal transition modulators, epigenetic mechanisms, hypoxia and cytokines. Identifying the exact molecular mechanisms of these dysregulated processes could help to discover the key targets for therapeutic purposes and utilizing them as diagnostic, prognostic and predictors of the clinical course of patients. In this review article, we describe different aspects of thyroid cancer metastasis by focusing on defective genes and pathways involved in its metastatic spread.
Collapse
Affiliation(s)
- Sadegh Rajabi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Heewa Shakib
- Cellular & Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Romina Dastmalchi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsoon Danesh-Afrooz
- Cellular & Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Karima
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular & Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
56
|
Connolly S, Newport D, McGourty K. The mechanical responses of advecting cells in confined flow. BIOMICROFLUIDICS 2020; 14:031501. [PMID: 32454924 PMCID: PMC7200165 DOI: 10.1063/5.0005154] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/21/2020] [Indexed: 05/03/2023]
Abstract
Fluid dynamics have long influenced cells in suspension. Red blood cells and white blood cells are advected through biological microchannels in both the cardiovascular and lymphatic systems and, as a result, are subject to a wide variety of complex fluidic forces as they pass through. In vivo, microfluidic forces influence different biological processes such as the spreading of infection, cancer metastasis, and cell viability, highlighting the importance of fluid dynamics in the blood and lymphatic vessels. This suggests that in vitro devices carrying cell suspensions may influence the viability and functionality of cells. Lab-on-a-chip, flow cytometry, and cell therapies involve cell suspensions flowing through microchannels of approximately 100-800 μ m. This review begins by examining the current fundamental theories and techniques behind the fluidic forces and inertial focusing acting on cells in suspension, before exploring studies that have investigated how these fluidic forces affect the reactions of suspended cells. In light of these studies' findings, both in vivo and in vitro fluidic cell microenvironments shall also be discussed before concluding with recommendations for the field.
Collapse
Affiliation(s)
- S Connolly
- School of Engineering, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | - D Newport
- School of Engineering, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland
| | | |
Collapse
|
57
|
Ishiguro S, Upreti D, Robben N, Burghart R, Loyd M, Ogun D, Le T, Delzeit J, Nakashima A, Thakkar R, Nakashima A, Suzuki K, Comer J, Tamura M. Water extract from Euglena gracilis prevents lung carcinoma growth in mice by attenuation of the myeloid-derived cell population. Biomed Pharmacother 2020; 127:110166. [PMID: 32361165 DOI: 10.1016/j.biopha.2020.110166] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/22/2022] Open
Abstract
The partially purified water extract from Euglena gracilis (EWE) was evaluated for its antitumor and immunomodulatory effects in cell cultures and in a mouse orthotopic lung carcinoma allograft model. In two-dimensional cell culture, the EWE treatment inhibited cell growth of both murine Lewis lung carcinoma (LLC) and human lung carcinoma cells (A549 and H1299) in a dose- and time-dependent manner. In contrast, the growth of mouse bone marrow cells (BMCs), but not mouse splenocytes (SPLs), was stimulated by the treatment with EWE. In three-dimensional spheroid culture, spheroid growth of LLC cells was significantly attenuated by EWE treatment. In a mouse LLC orthotopic allograft model, pretreatment with EWE (150-200 mg/kg/day, via drinking water) three weeks prior to the LLC cell inoculation, but not post-treatment after LLC cell inoculation, significantly attenuated the growth of LLC tumors in immunocompetent syngeneic mouse lung. This tumor growth attenuation coincided with a significant decrease in the population of myeloid-derived cells, primarily neutrophils. Flow cytometric analysis revealed that the EWE treatment significantly attenuated growth of granulocytic myeloid-derived suppressor cells (gMDSC) in BMCs and that this decrease was due to induction of gMDSC-specific apoptosis and differentiation of monocytic MDSCs (mMDSC) to macrophages. The present study provides evidence that EWE pretreatment inhibits lung carcinoma growth mainly by stimulating host antitumor immunity through attenuation of growth of gMDSCs and decreasing the number of peripheral granulocytes. This study suggests that the partially purified extract derived from Euglena gracilis contains significant bioactive materials that prevent lung carcinoma growth.
Collapse
Affiliation(s)
- Susumu Ishiguro
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Deepa Upreti
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Nicole Robben
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Riley Burghart
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Mayme Loyd
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Damilola Ogun
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Tran Le
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Jennifer Delzeit
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Arashi Nakashima
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Ravindra Thakkar
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | | | - Kengo Suzuki
- Euglena Co., Ltd., Minato-ku, Tokyo 108-0014, Japan.
| | - Jeffrey Comer
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| | - Masaaki Tamura
- Department of Anatomy & Physiology, Kansas State University College of Veterinary Medicine, Manhattan, KS 66506, United States.
| |
Collapse
|
58
|
The endothelial barrier and cancer metastasis: Does the protective facet of platelet function matter? Biochem Pharmacol 2020; 176:113886. [PMID: 32113813 DOI: 10.1016/j.bcp.2020.113886] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/24/2020] [Indexed: 12/16/2022]
Abstract
Overwhelming evidence suggests that platelets have a detrimental role in promoting cancer spread via platelet-cancer cell interactions linked to thrombotic mechanisms. On the other hand, a beneficial role of platelets in the preservation of the endothelial barrier in inflammatory conditions has been recently described, a phenomenon that could also operate in cancer-related inflammation. It is tempting to speculate that some antiplatelet strategies to combat cancer metastasis may impair the endogenous platelet-dependent mechanisms preserving endothelial barrier function. If the protective function of platelets is impaired, it may lead to increased endothelial permeability and more efficient cancer cell intravasation in the primary tumor and cancer cell extravasation at metastatic sites. In this commentary, we discuss current evidence that could support this hypothesis.
Collapse
|
59
|
Connolly S, McGourty K, Newport D. The in vitro inertial positions and viability of cells in suspension under different in vivo flow conditions. Sci Rep 2020; 10:1711. [PMID: 32015362 PMCID: PMC6997401 DOI: 10.1038/s41598-020-58161-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/31/2019] [Indexed: 12/21/2022] Open
Abstract
The influence of Poiseuille flow on cell viability has applications in the areas of cancer metastasis, lab-on-a-chip devices and flow cytometry. Indeed, retaining cell viability is important in the emerging field of cell therapy as cells need to be returned to patients’ bodies. Despite this, it is unclear how this fundamental fluid regime affects cell viability. This study investigated the influence that varying flow rate, and the corresponding wall shear stress (τw) has on the viability and inertial positions of circulating cells in laminar pipe flow. The viability of two representative cell lines under different shear stresses in two different systems were investigated while particle streak imaging was used to determine their inertial positions. It was found that peristaltic pumps have a negative effect on cell viability in comparison to syringe pumps. Increasing shear stress in a cone and plate above 3 Pa caused an increase in cell death, however, τw as high as 10 Pa in circulation has little to no effect on cell viability. Inertial lift forces that move cells towards the centre of the channel protect them from experiencing detrimental levels of τw, indicating that τw in Poiseuille flow is not a good predictor of cell viability during advection.
Collapse
Affiliation(s)
- Sinead Connolly
- School of Engineering, Bernal Institute, University of Limerick, Limerick, Ireland
| | - Kieran McGourty
- School of Natural Sciences, Bernal Institute, Health Research Institute, University of Limerick, Limerick, Ireland.
| | - David Newport
- School of Engineering, Bernal Institute, University of Limerick, Limerick, Ireland.
| |
Collapse
|
60
|
Di Liello R, Cimmino F, Simón S, Giunta EF, De Falco V, Martín-Martorell P. Role of liquid biopsy for thoracic cancers immunotherapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:183-199. [PMID: 36046196 PMCID: PMC9400760 DOI: 10.37349/etat.2020.00012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/11/2020] [Indexed: 01/09/2023] Open
Abstract
Immunotherapy has shifted the therapeutic landscape in thoracic cancers. However, assessment of biomarkers for patient selection and disease monitoring remain challenging, especially considering the lack of tissue sample availability for clinical and research purposes. In this scenario, liquid biopsy (LB), defined as the study and characterization of biomarkers in body fluids, represents a useful alternative strategy. In other malignancies such as colorectal cancer, breast cancer or melanoma, the potential of LB has been more extensively explored for monitoring minimal residual disease or response to treatment, and to investigate mechanisms of resistance to targeted agents. Even if various experiences have already been published about the applications of LB in immunotherapy in thoracic cancers, the standardization of methodology and assessment of its clinical utility is still pending. In this review, the authors will focus on the applications of LB in immunotherapy in non-small cell lung cancer, small cell lung cancer, and malignant pleural mesothelioma, describing available data and future perspectives.
Collapse
Affiliation(s)
- Raimondo Di Liello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy,Medical Oncology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain,Correspondence: Raimondo Di Liello, Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy.
| | | | - Soraya Simón
- Medical Oncology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Emilio Francesco Giunta
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Vincenzo De Falco
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Paloma Martín-Martorell
- Medical Oncology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| |
Collapse
|
61
|
Mechanics of actin filaments in cancer onset and progress. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:205-243. [DOI: 10.1016/bs.ircmb.2020.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
62
|
Wettschureck N, Strilic B, Offermanns S. Passing the Vascular Barrier: Endothelial Signaling Processes Controlling Extravasation. Physiol Rev 2019; 99:1467-1525. [PMID: 31140373 DOI: 10.1152/physrev.00037.2018] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A central function of the vascular endothelium is to serve as a barrier between the blood and the surrounding tissue of the body. At the same time, solutes and cells have to pass the endothelium to leave or to enter the bloodstream to maintain homeostasis. Under pathological conditions, for example, inflammation, permeability for fluid and cells is largely increased in the affected area, thereby facilitating host defense. To appropriately function as a regulated permeability filter, the endothelium uses various mechanisms to allow solutes and cells to pass the endothelial layer. These include transcellular and paracellular pathways of which the latter requires remodeling of intercellular junctions for its regulation. This review provides an overview on endothelial barrier regulation and focuses on the endothelial signaling mechanisms controlling the opening and closing of paracellular pathways for solutes and cells such as leukocytes and metastasizing tumor cells.
Collapse
Affiliation(s)
- Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Boris Strilic
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research , Bad Nauheim , Germany ; and Centre for Molecular Medicine, Medical Faculty, J.W. Goethe University Frankfurt , Frankfurt , Germany
| |
Collapse
|
63
|
Wall TP, Crowley PD, Sherwin A, Foley AG, Buggy DJ. Effects of Lidocaine and Src Inhibition on Metastasis in a Murine Model of Breast Cancer Surgery. Cancers (Basel) 2019; 11:cancers11101414. [PMID: 31546727 PMCID: PMC6826872 DOI: 10.3390/cancers11101414] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer recurs in 20% of patients following intended curative resection. In vitro data indicates that amide local anaesthetics, including lidocaine, inhibit cancer cell metastasis by inhibiting the tyrosine kinase enzyme Src. In a murine breast cancer surgery model, systemic lidocaine reduces postoperative pulmonary metastases. We investigated whether the additional administration of bosutinib (a known Src inhibitor) influences lidocaine’s observed beneficial effect in this in vivo model. Female BALB/c mice (n = 95) were inoculated with 25,000 4T1 cells into the mammary fad pad and after 7 days the resulting tumours were excised under sevoflurane anaesthesia. Experimental animals were randomized to one of four treatments administered intravenously prior to excision: lidocaine, bosutinib, both lidocaine and bosutinib in combination, or saline. Animals were euthanized 14 days post-surgery and lung and liver metastatic colonies were evaluated. Post-mortem serum was analysed for MMP-2 and MMP-9, pro-metastatic enzymes whose expression is influenced by the Src pathway. Lidocaine reduced lung, but not liver metastatic colonies versus sevoflurane alone (p = 0.041), but bosutinib alone had no metastasis-inhibiting effect. When combined with lidocaine, bosutinib reversed the anti-metastatic effect observed with lidocaine on sevoflurane anaesthesia. Only lidocaine alone reduced MMP-2 versus sevoflurane (p = 0.044). Both bosutinib (p = 0.001) and bosutinib/lidocaine combined (p = 0.001) reduced MMP-9 versus sevoflurane, whereas lidocaine alone did not. In a murine surgical breast cancer model, the anti-metastatic effects of lidocaine under sevoflurane anaesthesia are abolished by the Src inhibitor bosutinib, and lidocaine reduces serum MMP-2. These results suggest that lidocaine may act, at least partly, via an inhibitory effect on MMP-2 expression to reduce pulmonary metastasis, but whether this is due to an effect on Src or via another pathway remains unclear.
Collapse
Affiliation(s)
- Thomas P Wall
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 KH4C Dublin, Ireland.
- Conway Institute for Biomolecular and Biomedical Research, School of Medicine University College Dublin, D04 V1W8 Dublin, Ireland.
| | - Peter D Crowley
- Conway Institute for Biomolecular and Biomedical Research, School of Medicine University College Dublin, D04 V1W8 Dublin, Ireland.
| | - Aislinn Sherwin
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 KH4C Dublin, Ireland.
| | - Andrew G Foley
- Berand Neuropharmacology Ltd., NovaUCD, D04 V1W8 Dublin, Ireland.
| | - Donal J Buggy
- Department of Anaesthesiology & Perioperative Medicine, Mater University Hospital, School of Medicine, University College Dublin, D07 KH4C Dublin, Ireland.
- Conway Institute for Biomolecular and Biomedical Research, School of Medicine University College Dublin, D04 V1W8 Dublin, Ireland.
- Outcomes Research, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
64
|
Reddy GP, Reddy LV, Kim S. CANCER BIOLOGY AND PATHOLOGY. Cancer 2019. [DOI: 10.1002/9781119645214.ch2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
65
|
Investigating circulating tumor cells and distant metastases in patient-derived orthotopic xenograft models of triple-negative breast cancer. Breast Cancer Res 2019; 21:98. [PMID: 31462307 PMCID: PMC6714238 DOI: 10.1186/s13058-019-1182-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/05/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) represent a temporal "snapshot" of a patient's cancer and changes that occur during disease evolution. There is an extensive literature studying CTCs in breast cancer patients, and particularly in those with metastatic disease. In parallel, there is an increasing use of patient-derived models in preclinical investigations of human cancers. Yet studies are still limited demonstrating CTC shedding and metastasis formation in patient-derived models of breast cancer. METHODS We used seven patient-derived orthotopic xenograft (PDOX) models generated from triple-negative breast cancer (TNBC) patients to study CTCs and distant metastases. Tumor fragments from PDOX tissue from each of the seven models were implanted into 57 NOD scid gamma (NSG) mice, and tumor growth and volume were monitored. Human CTC capture from mouse blood was first optimized on the marker-agnostic Vortex CTC isolation platform, and whole blood was processed from 37 PDOX tumor-bearing mice. RESULTS Staining and imaging revealed the presence of CTCs in 32/37 (86%). The total number of CTCs varied between different PDOX tumor models and between individual mice bearing the same PDOX tumors. CTCs were heterogeneous and showed cytokeratin (CK) positive, vimentin (VIM) positive, and mixed CK/VIM phenotypes. Metastases were detected in the lung (20/57, 35%), liver (7/57, 12%), and brain (1/57, less than 2%). The seven different PDOX tumor models displayed varying degrees of metastatic potential, including one TNBC PDOX tumor model that failed to generate any detectable metastases (0/8 mice) despite having CTCs present in the blood of 5/5 tested, suggesting that CTCs from this particular PDOX tumor model may typify metastatic inefficiency. CONCLUSION PDOX tumor models that shed CTCs and develop distant metastases represent an important tool for investigating TNBC.
Collapse
|
66
|
Paul CD, Bishop K, Devine A, Paine EL, Staunton JR, Thomas SM, Thomas JR, Doyle AD, Miller Jenkins LM, Morgan NY, Sood R, Tanner K. Tissue Architectural Cues Drive Organ Targeting of Tumor Cells in Zebrafish. Cell Syst 2019; 9:187-206.e16. [PMID: 31445892 DOI: 10.1016/j.cels.2019.07.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 03/28/2019] [Accepted: 06/27/2019] [Indexed: 01/03/2023]
Abstract
Tumor cells encounter a myriad of physical cues upon arrest and extravasation in capillary beds. Here, we examined the role of physical factors in non-random organ colonization using a zebrafish xenograft model. We observed a two-step process by which mammalian mammary tumor cells showed non-random organ colonization. Initial homing was driven by vessel architecture, where greater numbers of cells became arrested in the topographically disordered blood vessels of the caudal vascular plexus (CVP) than in the linear vessels in the brain. Following arrest, bone-marrow- and brain-tropic clones exhibited organ-specific patterns of extravasation. Extravasation was mediated by β1 integrin, where knockdown of β1 integrin reduced extravasation in the CVP but did not affect extravasation of a brain-tropic clone in the brain. In contrast, silencing myosin 1B redirected early colonization from the brain to the CVP. Our results suggest that organ selectivity is driven by both vessel topography and cell-type-dependent extravasation.
Collapse
Affiliation(s)
- Colin D Paul
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kevin Bishop
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexus Devine
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elliott L Paine
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jack R Staunton
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sarah M Thomas
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joanna R Thomas
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew D Doyle
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lisa M Miller Jenkins
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Y Morgan
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20814, USA
| | - Raman Sood
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kandice Tanner
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
67
|
Onken JS, Fekonja LS, Wehowsky R, Hubertus V, Vajkoczy P. Metastatic dissemination patterns of different primary tumors to the spine and other bones. Clin Exp Metastasis 2019; 36:493-498. [PMID: 31420767 DOI: 10.1007/s10585-019-09987-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/09/2019] [Indexed: 11/30/2022]
Abstract
Metastatic spine disease (MSD) is a severe event in cancer patients. Experimental data indicate that bone metastasis is mostly mediated by blood flow-dependent, passive arrest of circulating tumor cells to the bone metastatic niche (BMN). Here, we have set out to test these experimental observations in a clinical, human setting to improve our understanding of MSD. 507 patients, treated on spinal metastases in our institution from 2005 to 2015 were retrospectively evaluated. We identified 259 patients with accessible staging reports of the skeleton before and at initial diagnosis of MSD. Data analysis comprised localizations of bone metastases, underlying malignancy and time to development of MSD. Dissemination pattern of bone metastasis was correlated with red bone marrow (RBM) content of the respective bone as a measure of blood flow. Spinal metastases occurred most frequently in lung cancer (21%), prostate cancer (19%), and breast cancer (12%). At the diagnosis of MSD, majority of patients have multiple extra-spinal bone metastases (2/3). The distribution of metastases to extra-spinal bones and to the spine is mostly proportional to the RBM content of the involved bone. Corresponding to the high RBM content, thoracic spine, pelvic bones and ribs represent a predilection site for bone metastasis. We confirm a distinct preference of cancer types to metastasize to bones. When it comes to bone metastases all primaries show uniform distribution pattern, which supports the hypothesis of a predominantly blood flow-dependent distribution of tumor cells and passive arrest to the BMN rather than a spine-specific homing mechanism.
Collapse
Affiliation(s)
- Julia S Onken
- Department of Neurosurgery, Universitätsmedizin Charite, Berlin, Germany
| | - Lucius S Fekonja
- Department of Neurosurgery, Universitätsmedizin Charite, Berlin, Germany
| | - Romy Wehowsky
- Department of Neurosurgery, Universitätsmedizin Charite, Berlin, Germany
| | - Vanessa Hubertus
- Department of Neurosurgery, Universitätsmedizin Charite, Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Universitätsmedizin Charite, Berlin, Germany. .,Neurochirurgische Klinik, Universitätsmedizin Charite, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
68
|
Kim H, Ishibashi K, Matsuo K, Kira A, Okada T, Watanabe K, Inada M, Nakamura C. Quantitative Measurements of Intercellular Adhesion Strengths between Cancer Cells with Different Malignancies Using Atomic Force Microscopy. Anal Chem 2019; 91:10557-10563. [DOI: 10.1021/acs.analchem.9b01569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Hyonchol Kim
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Kenta Ishibashi
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Kosuke Matsuo
- Product Development Center, Japan Aviation Electronics Ind., Ltd., 3-1-1 Musashino, Akishima, Tokyo 196-8555, Japan
| | - Atsushi Kira
- Product Development Center, Japan Aviation Electronics Ind., Ltd., 3-1-1 Musashino, Akishima, Tokyo 196-8555, Japan
| | - Tomoko Okada
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
| | - Kenta Watanabe
- Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Masaki Inada
- Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| | - Chikashi Nakamura
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan
- Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei, Tokyo 184-8588, Japan
| |
Collapse
|
69
|
Mollica H, Palomba R, Primavera R, Decuzzi P. Two-Channel Compartmentalized Microfluidic Chip for Real-Time Monitoring of the Metastatic Cascade. ACS Biomater Sci Eng 2019; 5:4834-4843. [DOI: 10.1021/acsbiomaterials.9b00697] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Hilaria Mollica
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, Via Morego 30, Genoa 16163, Italy
- DIBRIS, University of Genova, Via Opera Pia 13, Genoa 16145, Italy
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, Via Morego 30, Genoa 16163, Italy
| | - Rosita Primavera
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, Via Morego 30, Genoa 16163, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, Via Morego 30, Genoa 16163, Italy
| |
Collapse
|
70
|
Ingangi V, Minopoli M, Ragone C, Motti ML, Carriero MV. Role of Microenvironment on the Fate of Disseminating Cancer Stem Cells. Front Oncol 2019; 9:82. [PMID: 30847298 PMCID: PMC6393337 DOI: 10.3389/fonc.2019.00082] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/29/2019] [Indexed: 12/12/2022] Open
Abstract
Disseminating Cancer Stem Cells (CSCs) initiate growth in specific niches of the host tissues, the cellular and molecular components of which sustain signaling pathways that support their survival, self-renewal dormancy and reactivation. In the metastatic niche, tumor cells may enter in a dormant state to survive and, consequently, the metastasis can remain latent for years. Despite the clinical importance of metastatic latency, little is known about what induces CSCs to enter a dormant state and what allows them to remain viable for years in this state. CSCs exhibit genetic, epigenetic and cellular adaptations that confer resistance to classical therapeutic approaches. The identification of potential CSC targets is complicated by the fact that CSCs may arise as a consequence of their relationship with the local microenvironment into the metastatic niches. Indeed, microenvironment modulates the capability of CSCs to evade the innate immune response and survive. Some new therapeutic options that include drugs targeting microenvironment components are achieving encouraging results in reducing the number of CSCs in tumors and/or overcoming their resistance in preclinical studies. This review will focus on specific CSC features with an emphasis on the role of tumor microenvironment in supporting metastatic dissemination of CSCs. In addition, it sheds light on potential microenvironment-targeted therapies aimed to counteract seeding and survival of CSCs in the metastatic niche.
Collapse
Affiliation(s)
- Vincenzo Ingangi
- IRCCS Istituto Nazionale Tumori, Fondazione G. Pascale, Naples, Italy
| | - Michele Minopoli
- IRCCS Istituto Nazionale Tumori, Fondazione G. Pascale, Naples, Italy
| | - Concetta Ragone
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Maria Letizia Motti
- IRCCS Istituto Nazionale Tumori, Fondazione G. Pascale, Naples, Italy.,Department of Sport Science and Wellness, University Parthenope, Naples, Italy
| | | |
Collapse
|
71
|
Villanueva F, Araya H, Briceño P, Varela N, Stevenson A, Jerez S, Tempio F, Chnaiderman J, Perez C, Villarroel M, Concha E, Khani F, Thaler R, Salazar-Onfray F, Stein GS, van Wijnen AJ, Galindo M. The cancer-related transcription factor RUNX2 modulates expression and secretion of the matricellular protein osteopontin in osteosarcoma cells to promote adhesion to endothelial pulmonary cells and lung metastasis. J Cell Physiol 2019; 234:13659-13679. [PMID: 30637720 DOI: 10.1002/jcp.28046] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 12/06/2018] [Indexed: 12/19/2022]
Abstract
Osteosarcomas are bone tumors that frequently metastasize to the lung. Aberrant expression of the transcription factor, runt-related transcription factor 2 (RUNX2), is a key pathological feature in osteosarcoma and associated with loss of p53 and miR-34 expression. Elevated RUNX2 may transcriptionally activate genes mediating tumor progression and metastasis, including the RUNX2 target gene osteopontin (OPN/SPP1). This gene encodes a secreted matricellular protein produced by osteoblasts to regulate bone matrix remodeling and tissue calcification. Here we investigated whether and how the RUNX2/OPN axis regulates lung metastasis of osteosarcoma. Importantly, RUNX2 depletion attenuates lung metastasis of osteosarcoma cells in vivo. Using next-generation RNA-sequencing, protein-based assays, as well as the loss- and gain-of-function approaches in selected osteosarcoma cell lines, we show that osteopontin messenger RNA levels closely correlate with RUNX2 expression and that RUNX2 controls the levels of secreted osteopontin. Elevated osteopontin levels promote heterotypic cell-cell adhesion of osteosarcoma cells to human pulmonary microvascular endothelial cells, but not in the presence of neutralizing antibodies. Collectively, these findings indicate that the RUNX2/OPN axis regulates the ability of osteosarcoma cells to attach to pulmonary endothelial cells as a key step in metastasis of osteosarcoma cells to the lung.
Collapse
Affiliation(s)
- Francisco Villanueva
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Hector Araya
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pedro Briceño
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Nelson Varela
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Department of Medical Technology, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Andres Stevenson
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sofia Jerez
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fabian Tempio
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Jonas Chnaiderman
- Program of Virology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Carola Perez
- Laboratory Animal Facility, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Milena Villarroel
- Department of Oncology, Hospital Dr. Luis Calvo Mackenna, Santiago, Chile.,National Child Programme of Antineoplastic Drugs (PINDA), Santiago, Chile
| | - Emma Concha
- Department of Oncology, Hospital Dr. Luis Calvo Mackenna, Santiago, Chile
| | - Farzaneh Khani
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Flavio Salazar-Onfray
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Gary S Stein
- Department of Biochemistry, University of Vermont Cancer Center, The Robert Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mario Galindo
- Millennium Institute on Immunology and Immunotherapy, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
72
|
Mariscal J, Fernandez-Puente P, Calamia V, Abalo A, Santacana M, Matias-Guiu X, Lopez-Lopez R, Gil-Moreno A, Alonso-Alconada L, Abal M. Proteomic Characterization of Epithelial-Like Extracellular Vesicles in Advanced Endometrial Cancer. J Proteome Res 2019; 18:1043-1053. [PMID: 30585730 DOI: 10.1021/acs.jproteome.8b00750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Endometrial cancer (EC) is the most frequent gynecological cancer. Tumor dissemination affecting ∼20% of EC patients is characterized at the primary carcinoma by epithelial-to-mesenchymal transition (EMT) associated with myometrial infiltration. At distant sites, the interaction of circulating tumor cells (CTCs) with the microenvironment is crucial for metastatic colonization, with a participation of the extracellular vesicles (EVs). We comprehensively approached these primary and secondary sites to study the impact of tumor EVs on the metastatic efficiency of CTCs in EC. Tumor EVs in circulation reproduce the epithelial phenotype predominant in the primary carcinoma, whereas CTCs are characterized by an EMT phenotype. We modeled this EMT-related clinical scenario in the Hec1A endometrial cell line and characterized the epithelial-like EVs in circulation by SILAC proteome analysis. The identification of proteins involved in cell-cell and cell-matrix interaction and binding, together with in vitro evidence of an improved adhesion of CTC to a functionalized endothelium, suggests a contribution of the epithelial-like EVs in the homing of CTCs at metastatic sites. Accordingly, adhesion protein LGALS3BP was found to be significantly enriched in circulating EVs from a cohort of EC patients with a high risk of recurrence by targeted proteomics (multiple reaction monitoring), highlighting its potential in liquid biopsy in EC.
Collapse
Affiliation(s)
- Javier Mariscal
- Translational Medical Oncology, CIBERONC, Health Research Institute of Santiago (IDIS) , University Hospital of Santiago (SERGAS) , Santiago de Compostela 15706 , Spain
| | - Patricia Fernandez-Puente
- Proteomics Group-PBR2-ProteoRed/ISCIII, Rheumatology Division, Instituto de Investigación Biomédica de A Coruña (INIBIC) , University Hospital A Coruña (SERGAS), Universidade da Coruña , A Coruña 15006 , Spain
| | - Valentina Calamia
- Proteomics Group-PBR2-ProteoRed/ISCIII, Rheumatology Division, Instituto de Investigación Biomédica de A Coruña (INIBIC) , University Hospital A Coruña (SERGAS), Universidade da Coruña , A Coruña 15006 , Spain
| | - Alicia Abalo
- Translational Medical Oncology, CIBERONC, Health Research Institute of Santiago (IDIS) , University Hospital of Santiago (SERGAS) , Santiago de Compostela 15706 , Spain
| | - Maria Santacana
- Hospital Universitari Arnau de Vilanova , University of Lleida, IRBLLEIDA, CIBERONC , Lleida 08080 , Spain
| | - Xavier Matias-Guiu
- Hospital Universitari Arnau de Vilanova , University of Lleida, IRBLLEIDA, CIBERONC , Lleida 08080 , Spain
| | - Rafael Lopez-Lopez
- Translational Medical Oncology, CIBERONC, Health Research Institute of Santiago (IDIS) , University Hospital of Santiago (SERGAS) , Santiago de Compostela 15706 , Spain
| | - Antonio Gil-Moreno
- Biomedical Research Group in Gynecology, Vall Hebron Research Institute (VHIR) , Universitat Autonoma de Barcelona, CIBERONC , Barcelona 08035 , Spain
| | - Lorena Alonso-Alconada
- Translational Medical Oncology, CIBERONC, Health Research Institute of Santiago (IDIS) , University Hospital of Santiago (SERGAS) , Santiago de Compostela 15706 , Spain
| | - Miguel Abal
- Translational Medical Oncology, CIBERONC, Health Research Institute of Santiago (IDIS) , University Hospital of Santiago (SERGAS) , Santiago de Compostela 15706 , Spain
| |
Collapse
|
73
|
Li L, Zhang J, Jiang X, Li Q. Promising clinical application of ctDNA in evaluating immunotherapy efficacy. Am J Cancer Res 2018; 8:1947-1956. [PMID: 30416847 PMCID: PMC6220137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/24/2018] [Indexed: 06/09/2023] Open
Abstract
An increasing number of promising immunotherapies and related clinical trials have led to several major breakthroughs in multiple cancers, but a reliable and precise biomarker for evaluating efficacy and prognosis has not yet been established. As a typical representation of a liquid biopsy, circulating cell-free DNA (ctDNA) possesses the functions and advantages of tissue biopsy but its distinct advantages of convenience, real-time nature, non-invasiveness and homogeneity make it superior to tissue biopsy. Indeed, compared with routine imaging and tumor markers, ctDNA offers an earlier indication and provides more precise information. ctDNA is reportedly able to identify immunotherapy responders, evaluate efficacy and survival time, screen immune checkpoint inhibitor resistance and pseudo-progress and predict tumor recurrence and metastasis. Thus, ctDNA can act as an "Eagle Eye" by comprehensively monitoring both macro- and micro-changes in the immunotherapy process. Although ctDNA has become a research topic of interest, its limitations cannot be ignored, and improvements in its sensitivity and standardization are urgently needed. This review reveals the advantages and limitations of ctDNA as a precise biomarker and supports the feasibility of using ctDNA detection for common monitoring during immunotherapy.
Collapse
Affiliation(s)
- Li Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
| | - Jun Zhang
- Department of Hematopathology, University of Texas MD Anderson Cancer CenterHouston, TX 77030, US
| | - Xiaoyue Jiang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical UniversityBeijing 100050, China
| |
Collapse
|
74
|
Vilhav C, Engström C, Naredi P, Novotny A, Bourghardt-Fagman J, Iresjö BM, Asting AG, Lundholm K. Fractional uptake of circulating tumor cells into liver-lung compartments during curative resection of periampullary cancer. Oncol Lett 2018; 16:6331-6338. [PMID: 30405768 PMCID: PMC6202519 DOI: 10.3892/ol.2018.9435] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/02/2018] [Indexed: 12/25/2022] Open
Abstract
Circulating tumor cells (CTCs) are able to predict outcome in patients with breast, colon and prostate cancer and appear to be promising biomarkers of pancreatic carcinoma. The aim of the present study was to demonstrate a statistically significant portal-arterial difference of CTCs during curative resection of periampullary cancer. A commercially available instrument (IsofluxR) was used to quantify blood content of CTC in 10 patients with periampullary cancer according to preoperative diagnostics. Portal and arterial blood samples (~8 ml each) were simultaneously collected intra-operatively following surgical dissection prior to division of the pancreas for tumor removal. Quantitative CTC analyses were performed according to standardized protocols for immune-magnetic enrichment of CTC. Flow cytometry was applied for qualitative evaluations of various CTC markers in 7 patients. There was a statistically significant difference in the number of CTCs collected in the portal blood [58±14 cells per 100 ml; mean ± standard error (SE)] vs. arterial blood [24±7 cells per 100 ml (SE), P<0.025]. A fractional uptake of ≥40% across liver and lung compartments of assumed malignant CTC was estimated to correspond to the appearance of ~410 tumor cells per minute during pancreatic resections based on estimated hepatic blood flow, measured tumor cell mass and tumor cell proliferation activity. Complications in the collection of portal blood were not observed. A significant uptake across liver or lung compartments of potentially malignant tumor CTCs from periampullary carcinoma may represent a model to capture, define and characterize cell clones with metastatic potential in liver and lung tissues following surgical resection.
Collapse
Affiliation(s)
- Caroline Vilhav
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Cecilia Engström
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Ann Novotny
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Johan Bourghardt-Fagman
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Britt-Marie Iresjö
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Annika G Asting
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| | - Kent Lundholm
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg SE-41346, Sweden
| |
Collapse
|
75
|
Stojak M, Kaczara P, Motterlini R, Chlopicki S. Modulation of cellular bioenergetics by CO-releasing molecules and NO-donors inhibits the interaction of cancer cells with human lung microvascular endothelial cells. Pharmacol Res 2018; 136:160-171. [PMID: 30196104 DOI: 10.1016/j.phrs.2018.09.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/05/2018] [Indexed: 10/28/2022]
Abstract
Interactions between cancer cells and the endothelium play a crucial role during metastasis. Here we examined the effects of a carbon monoxide-releasing molecule (CORM-401) and a nitric oxide donor (PAPA NONOate) given alone or in combination on breast cancer cell adhesion and transmigration across the lung microvascular endothelium. We further explored whether the effects of CO and NO on cancer-endothelial cells interactions are linked with changes in cellular bioenergetics in breast cancer or endothelial cells. We found that CORM-401 and PAPA NONOate alone or in combination markedly decreased transmigration of breast cancer cells across human lung microvascular endothelial cells (hLMVEC), while cancer cell adhesion to the endothelium was diminished only by a combination of the two compounds. In hLMVECs, CORM-401 decreased glycolysis and stimulated mitochondrial respiration, while in breast cancer cells CORM-401 decreased both glycolysis and mitochondrial respiration. In contrast, PAPA NONOate decreased mitochondrial respiration and slightly stimulated glycolysis in both cell lines. When both donors were given together, mitochondrial respiration and glycolysis were both profoundly inhibited, and cancer-endothelial cells interactions were additively suppressed. Intercellular adhesion molecule-1 (ICAM-1), involved in breast cancer cell adhesion to hLMVECs, was downregulated by CORM-401 and PAPA NONOate, when applied alone, while a combination of both compounds did not cause any enhancement of ICAM-1 downregulation. In conclusion, our findings demonstrate that CO and NO differently affect cellular bioenergetics of cancer and endothelial cells and suggest that this phenomenon may contribute to additive anti-adhesive and anti-transmigratory effects of CO and NO. Pharmacological attenuation of metabolism represents a novel, effective way to prevent cancer cell interactions with the endothelium, that is an energy-demanding process.
Collapse
Affiliation(s)
- Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Patrycja Kaczara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Roberto Motterlini
- INSERM Unit 955, Equipe 12, Faculty of Medicine, University Paris-Est, Créteil, France
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland; Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
76
|
Ishikane S, Hosoda H, Nojiri T, Tokudome T, Mizutani T, Miura K, Akitake Y, Kimura T, Imamichi Y, Kawabe S, Toyohira Y, Yanagihara N, Takahashi-Yanaga F, Miyazato M, Miyamoto K, Kangawa K. Angiotensin II promotes pulmonary metastasis of melanoma through the activation of adhesion molecules in vascular endothelial cells. Biochem Pharmacol 2018; 154:136-147. [DOI: 10.1016/j.bcp.2018.04.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/12/2018] [Indexed: 12/22/2022]
|
77
|
Sellers ZP, Schneider G, Bujko K, Suszynska M, Pedziwiatr D. Do Cancer Cell Lines Have Fixed or Fluctuating Stem Cell Phenotypes? - Studies with the NTera2 Cell Line. Stem Cell Rev Rep 2018. [PMID: 28624968 DOI: 10.1007/s12015-017-9743-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
One of the important questions when studying established cancer cell lines is whether such cells contain a subpopulation of primitive cancer stem cells that maintains the expansion of the cell line. To address this issue, we performed studies on the established human embryonal carcinoma cell line NTera2 by evaluating the potential stemness of cells sorted according to their expression of the cell surface stem cell markers CD133 and SSEA4. By performing in vitro and in vivo assays, we observed different properties of cells expressing both, one, or neither of these antigens. While sorted SSEA4+ subpopulations exhibited the greatest propensity for migration toward normal serum and the highest seeding efficiency in the lungs of immunodeficient mice, CD133-SSEA4- cells displayed high seeding efficiency to the bone marrow after injection in vivo. It is worth noting that these properties did not depend on the size of the evaluated cells. To address the question of whether cancer stem cell phenotypes in cell lines are fixed or fluctuating, we sorted single cells according to their expression of CD133 and SSEA4 antigens and observed that cells which did not express these cancer stem cell markers gave rise to cells that express these markers after expansion in vitro. Therefore, our results support the idea that within established cancer cell lines, the phenotype of the cell subpopulation expressing cancer stem cell markers is not fixed but fluctuates during cell line expansion, and cells negative for these markers may acquire their expression.
Collapse
Affiliation(s)
- Zachariah P Sellers
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA.
| | - Gabriela Schneider
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Kamila Bujko
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Malwina Suszynska
- Stem Cell Institute at the James Graham Brown Cancer Center, University of Louisville, Louisville, KY, 40202, USA
| | - Daniel Pedziwiatr
- Department of Regenerative Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
78
|
Hemodynamic Forces Tune the Arrest, Adhesion, and Extravasation of Circulating Tumor Cells. Dev Cell 2018; 45:33-52.e12. [PMID: 29634935 DOI: 10.1016/j.devcel.2018.02.015] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/05/2018] [Accepted: 02/16/2018] [Indexed: 02/07/2023]
Abstract
Metastatic seeding is driven by cell-intrinsic and environmental cues, yet the contribution of biomechanics is poorly known. We aim to elucidate the impact of blood flow on the arrest and the extravasation of circulating tumor cells (CTCs) in vivo. Using the zebrafish embryo, we show that arrest of CTCs occurs in vessels with favorable flow profiles where flow forces control the adhesion efficacy of CTCs to the endothelium. We biophysically identified the threshold values of flow and adhesion forces allowing successful arrest of CTCs. In addition, flow forces fine-tune tumor cell extravasation by impairing the remodeling properties of the endothelium. Importantly, we also observe endothelial remodeling at arrest sites of CTCs in mouse brain capillaries. Finally, we observed that human supratentorial brain metastases preferably develop in areas with low perfusion. These results demonstrate that hemodynamic profiles at metastatic sites regulate key steps of extravasation preceding metastatic outgrowth.
Collapse
|
79
|
Pang K, Xie C, Yang Z, Suo Y, Zhu X, Wei D, Weng X, Wei X, Gu Z. Monitoring circulating prostate cancer cells by in vivo flow cytometry assesses androgen deprivation therapy on metastasis. Cytometry A 2018; 93:517-524. [PMID: 29683554 DOI: 10.1002/cyto.a.23369] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/28/2018] [Accepted: 03/07/2018] [Indexed: 01/08/2023]
Abstract
It remains controversial whether surgical castration prolongs survival rate and improves therapy prospects in patients suffering from prostate cancer. We used PC3 cell line to establish prostate tumor models. In vivo flow cytometry and ultrasonic imaging were used to monitor the process of prostate cancer growth, development and metastasis. We found out that the number of circulating tumor cells (CTCs) in orthotopic tumor model was higher than that in subcutaneous tumor model. The CTC number in orthotopic tumor model was due to burst growth, while CTC number in subcutaneous tumor model showed a gradual increase with tumor size. After androgen deprivation therapy (ADT) through testicular extraction, we constructed GFP-PC3 subcutaneous tumor models and orthotopic tumor models. We found dramatically decreased CTC number, relieved symptoms caused by the tumor, and significantly prolonged survival time after testicular extraction in orthotopically transplanted prostate tumor model, while the carcinogenesis process and metastases were little influenced by ADT in subcutaneous tumor model. ADT treatment can restrict tumor growth, decrease the CTC number significantly and inhibit distant invasion through inhibition of tumor proliferation and tumor angiogenesis in orthotopical prostate tumor model. © 2018 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Kai Pang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chengying Xie
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhangru Yang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.,Radiation Oncology Center, Fudan University Shanghai Cancer Center (FUSCC), Shanghai 200032, China
| | - Yuanzhen Suo
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xi Zhu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Dan Wei
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xiaofu Weng
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xunbin Wei
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, 3688 Nanhai Road, Shenzhen 518060, China
| | - Zhengqin Gu
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| |
Collapse
|
80
|
Rezaeeyan H, Shirzad R, McKee TD, Saki N. Role of chemokines in metastatic niche: new insights along with a diagnostic and prognostic approach. APMIS 2018; 126:359-370. [PMID: 29676815 DOI: 10.1111/apm.12818] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 01/04/2018] [Indexed: 01/10/2023]
Abstract
Chemokines are cytokines that are involved in the movement of leukocytes and the occurrence of immune responses. It has recently been noted that these cytokines play a role in the movement of cancer cells to different parts of the body and create a suitable environment [i.e. (pre) metastatic niche] for their growth and proliferation. We studied the role of chemokines in the metastasis of cancer cells, as well as their involvement in the proliferation and growth of these cells. Relevant literature was identified by a PubMed search (2005-2017) of English language papers using the terms 'chemokine,' 'metastasis niche,' and 'organotropism.' Based on the nature of cancer cells, the expression of chemokine receptors on these cells leads to metastasis to various organs, which ultimately causes changes in different signaling pathways. Finally, the targeting of chemokines on cancer cells could prevent the metastasis of cancer cells toward different organs.
Collapse
Affiliation(s)
- Hadi Rezaeeyan
- Research Center of Thalassemia & Hemoglobinopathy, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Shirzad
- WHO-Collaborating Centre for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Trevor D McKee
- Princess Margaret Cancer Centre, STTARR Innovation Facility, Toronto, ON, Canada
| | - Najmaldin Saki
- Research Center of Thalassemia & Hemoglobinopathy, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
81
|
Salehi B, Zucca P, Sharifi-Rad M, Pezzani R, Rajabi S, Setzer WN, Varoni EM, Iriti M, Kobarfard F, Sharifi-Rad J. Phytotherapeutics in cancer invasion and metastasis. Phytother Res 2018; 32:1425-1449. [DOI: 10.1002/ptr.6087] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/11/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Bahare Salehi
- Medical Ethics and Law Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Paolo Zucca
- Department of Biomedical Sciences; University of Cagliari; Cagliari Italy
| | - Mehdi Sharifi-Rad
- Department of Medical Parasitology; Zabol University of Medical Sciences; Zabol 61663-335 Iran
| | - Raffaele Pezzani
- OU Endocrinology, Dept. Medicine (DIMED); University of Padova; via Ospedale 105 Padova 35128 Italy
- AIROB, Associazione Italiana per la Ricerca Oncologica di Base; Padova Italy
| | - Sadegh Rajabi
- Department of Clinical Biochemistry, School of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - William N. Setzer
- Department of Chemistry; University of Alabama in Huntsville; Huntsville AL 35899 USA
| | - Elena Maria Varoni
- Department of Biomedical, Surgical and Dental Sciences; Milan State University; Milan Italy
| | - Marcello Iriti
- Department of Agricultural and Environmental Sciences; Milan State University; Milan Italy
| | - Farzad Kobarfard
- Phytochemistry Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Medicinal Chemistry, School of Pharmacy; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Javad Sharifi-Rad
- Phytochemistry Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Chemistry, Richardson College for the Environmental Science Complex; The University of Winnipeg; Winnipeg MB Canada
| |
Collapse
|
82
|
Chen W, Hoffmann AD, Liu H, Liu X. Organotropism: new insights into molecular mechanisms of breast cancer metastasis. NPJ Precis Oncol 2018; 2:4. [PMID: 29872722 PMCID: PMC5871901 DOI: 10.1038/s41698-018-0047-0] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 02/08/2023] Open
Abstract
Metastasis accounts for 90% of breast cancer mortality. Despite the significant progress made over the past decade in cancer medicine our understanding of metastasis remains limited, therefore preventing and targeting metastasis is not yet possible. Breast cancer cells preferentially metastasize to specific organs, known as “organotropic metastasis”, which is regulated by subtypes of breast cancer, host organ microenvironment, and cancer cells-organ interactions. The cross-talk between cancer cells and host organs facilitates the formation of the premetastatic niche and is augmented by factors released from cancer cells prior to the cancer cells’ arrival at the host organ. Moreover, host microenvironment and specific organ structure influence metastatic niche formation and interactions between cancer cells and local resident cells, regulating the survival of cancer cells and formation of metastatic lesions. Understanding the molecular mechanisms of organotropic metastasis is essential for biomarker-based prediction and prognosis, development of innovative therapeutic strategy, and eventual improvement of patient outcomes. In this review, we summarize the molecular mechanisms of breast cancer organotropic metastasis by focusing on tumor cell molecular alterations, stemness features, and cross-talk with the host environment. In addition, we also update some new progresses on our understanding about genetic and epigenetic alterations, exosomes, microRNAs, circulating tumor cells and immune response in breast cancer organotropic metastasis.
Collapse
Affiliation(s)
- Wenjing Chen
- 1Department of Pharmacology, Northwestern University, Chicago, IL USA
| | - Andrew D Hoffmann
- 1Department of Pharmacology, Northwestern University, Chicago, IL USA
| | - Huiping Liu
- 1Department of Pharmacology, Northwestern University, Chicago, IL USA.,2Department of Medicine, Division of Hematology and Oncology, Northwestern University, Chicago, IL USA.,3Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL USA
| | - Xia Liu
- 1Department of Pharmacology, Northwestern University, Chicago, IL USA.,3Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL USA
| |
Collapse
|
83
|
Chan JY, Ahmad Kayani AB, Md Ali MA, Kok CK, Yeop Majlis B, Hoe SLL, Marzuki M, Khoo ASB, Ostrikov K(K, Ataur Rahman M, Sriram S. Dielectrophoresis-based microfluidic platforms for cancer diagnostics. BIOMICROFLUIDICS 2018; 12:011503. [PMID: 29531634 PMCID: PMC5825230 DOI: 10.1063/1.5010158] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/27/2017] [Indexed: 05/15/2023]
Abstract
The recent advancement of dielectrophoresis (DEP)-enabled microfluidic platforms is opening new opportunities for potential use in cancer disease diagnostics. DEP is advantageous because of its specificity, low cost, small sample volume requirement, and tuneable property for microfluidic platforms. These intrinsic advantages have made it especially suitable for developing microfluidic cancer diagnostic platforms. This review focuses on a comprehensive analysis of the recent developments of DEP enabled microfluidic platforms sorted according to the target cancer cell. Each study is critically analyzed, and the features of each platform, the performance, added functionality for clinical use, and the types of samples, used are discussed. We address the novelty of the techniques, strategies, and design configuration used in improving on existing technologies or previous studies. A summary of comparing the developmental extent of each study is made, and we conclude with a treatment of future trends and a brief summary.
Collapse
Affiliation(s)
- Jun Yuan Chan
- Center for Advanced Materials and Green Technology, Multimedia University, 75450 Melaka, Malaysia
| | | | - Mohd Anuar Md Ali
- Institute of Microengineering and Nanoelectronics, Universiti Kebangsaan Malaysia, Bangi, 43600 Selangor, Malaysia
| | - Chee Kuang Kok
- Center for Advanced Materials and Green Technology, Multimedia University, 75450 Melaka, Malaysia
| | - Burhanuddin Yeop Majlis
- Institute of Microengineering and Nanoelectronics, Universiti Kebangsaan Malaysia, Bangi, 43600 Selangor, Malaysia
| | - Susan Ling Ling Hoe
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, 50588 Kuala Lumpur, Malaysia
| | - Marini Marzuki
- Molecular Pathology Unit, Cancer Research Centre, Institute for Medical Research, 50588 Kuala Lumpur, Malaysia
| | | | | | - Md. Ataur Rahman
- Functional Materials and Microsystems Research Group, Micro Nano Research Facility, RMIT University, Melbourne, Victoria 3001, Australia
| | - Sharath Sriram
- Functional Materials and Microsystems Research Group, Micro Nano Research Facility, RMIT University, Melbourne, Victoria 3001, Australia
| |
Collapse
|
84
|
Liu Q, Zhang H, Jiang X, Qian C, Liu Z, Luo D. Factors involved in cancer metastasis: a better understanding to "seed and soil" hypothesis. Mol Cancer 2017; 16:176. [PMID: 29197379 PMCID: PMC5712107 DOI: 10.1186/s12943-017-0742-4] [Citation(s) in RCA: 213] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023] Open
Abstract
Metastasis has intrigued researchers for more than 100 years. Despite the development of technologies and therapeutic strategies, metastasis is still the major cause of cancer-related death until today. The famous "seed and soil" hypothesis is widely cited and accepted, and it still provides significant instructions in cancer research until today. To our knowledge, there are few reviews that comprehensively and correlatively focus on both the seed and soil factors involved in cancer metastasis; moreover, despite the fact that increasingly underlying mechanisms and concepts have been defined recently, previous perspectives are appealing but may be limited. Hence, we reviewed factors involved in cancer metastasis, including both seed and soil factors. By integrating new concepts with the classic hypothesis, we aim to provide a comprehensive understanding of the "seed and soil" hypothesis and to conceptualize the framework for understanding factors involved in cancer metastasis. Based on a dynamic overview of this field, we also discuss potential implications for future research and clinical therapeutic strategies.
Collapse
Affiliation(s)
- Qiang Liu
- First Clinical Medical College, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Hongfei Zhang
- Queen Mary School, School of Medicine, Nanchang University, Nanchang, People's Republic of China
| | - Xiaoli Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China
| | - Caiyun Qian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang, Bayi Road, No.461, 330006, Nanchang, People's Republic of China.
| |
Collapse
|
85
|
Caballero D, Kaushik S, Correlo V, Oliveira J, Reis R, Kundu S. Organ-on-chip models of cancer metastasis for future personalized medicine: From chip to the patient. Biomaterials 2017; 149:98-115. [DOI: 10.1016/j.biomaterials.2017.10.005] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/15/2017] [Accepted: 10/02/2017] [Indexed: 02/09/2023]
|
86
|
Dexmedetomidine promotes metastasis in rodent models of breast, lung, and colon cancers. Br J Anaesth 2017; 120:188-196. [PMID: 29397129 DOI: 10.1016/j.bja.2017.11.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 08/04/2017] [Accepted: 09/03/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Perioperative strategies can significantly influence long-term cancer outcomes. Dexmedetomidine, an α2-adrenoceptor agonist, is increasingly used perioperatively for its sedative, analgesic, anxiolytic, and sympatholytic effects. Such actions might attenuate the perioperative promotion of metastases, but other findings suggest opposite effects on primary tumour progression. We tested the effects of dexmedetomidine in clinically relevant models of dexmedetomidine use on cancer metastatic progression. METHODS Dexmedetomidine was given to induce sub-hypnotic to sedative effects for 6-12 h, and its effects on metastasis formation, using various cancer types, were studied in naïve animals and in the context of stress and surgery. RESULTS Dexmedetomidine increased tumour-cell retention and growth of metastases of a mammary adenocarcinoma (MADB 106) in F344 rats, Lewis lung carcinoma (3LL) in C57BL/6 mice, and colon adenocarcinoma (CT26) in BALB/c mice. The metastatic burden increased in both sexes and in all organs tested, including lung, liver, and kidney, as well as in brain employing a novel external carotid-artery inoculation approach. These effects were mediated through α2-adrenergic, but not α1-adrenergic, receptors. Low sub-hypnotic doses of dexmedetomidine were moderately beneficial in attenuating the deleterious effects of one stress paradigm, but not of the surgery or other stressors. CONCLUSIONS The findings call for mechanistic translational studies to understand these deleterious effects of dexmedetomidine, and warrant prospective clinical trials to assess the impact of perioperative dexmedetomidine use on outcomes in cancer patients.
Collapse
|
87
|
Harlepp S, Thalmann F, Follain G, Goetz JG. Hemodynamic forces can be accurately measured in vivo with optical tweezers. Mol Biol Cell 2017; 28:3252-3260. [PMID: 28904205 PMCID: PMC5687027 DOI: 10.1091/mbc.e17-06-0382] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/15/2022] Open
Abstract
Force sensing and generation at the tissue and cellular scale is central to many biological events. There is a growing interest in modern cell biology for methods enabling force measurements in vivo. Optical trapping allows noninvasive probing of piconewton forces and thus emerged as a promising mean for assessing biomechanics in vivo. Nevertheless, the main obstacles lie in the accurate determination of the trap stiffness in heterogeneous living organisms, at any position where the trap is used. A proper calibration of the trap stiffness is thus required for performing accurate and reliable force measurements in vivo. Here we introduce a method that overcomes these difficulties by accurately measuring hemodynamic profiles in order to calibrate the trap stiffness. Doing so, and using numerical methods to assess the accuracy of the experimental data, we measured flow profiles and drag forces imposed to trapped red blood cells of living zebrafish embryos. Using treatments enabling blood flow tuning, we demonstrated that such a method is powerful in measuring hemodynamic forces in vivo with accuracy and confidence. Altogether this study demonstrates the power of optical tweezing in measuring low range hemodynamic forces in vivo and offers an unprecedented tool in both cell and developmental biology.
Collapse
Affiliation(s)
- Sébastien Harlepp
- Université de Strasbourg, 67000 Strasbourg, France .,IPCMS, UMR7504, 67200 Strasbourg, France.,LabEx NIE, Université de Strasbourg, 67000 Strasbourg, France
| | - Fabrice Thalmann
- Université de Strasbourg, 67000 Strasbourg, France.,ICS, UPR22, 67034 Strasbourg, France
| | - Gautier Follain
- Université de Strasbourg, 67000 Strasbourg, France.,Inserm UMR_S1109, MN3T, 67200 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, 67000 Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| | - Jacky G Goetz
- Université de Strasbourg, 67000 Strasbourg, France .,Inserm UMR_S1109, MN3T, 67200 Strasbourg, France.,LabEx Medalis, Université de Strasbourg, 67000 Strasbourg, France.,Fédération de Médecine Translationnelle de Strasbourg (FMTS), 67000 Strasbourg, France
| |
Collapse
|
88
|
Strilic B, Offermanns S. Intravascular Survival and Extravasation of Tumor Cells. Cancer Cell 2017; 32:282-293. [PMID: 28898694 DOI: 10.1016/j.ccell.2017.07.001] [Citation(s) in RCA: 279] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/24/2017] [Accepted: 07/05/2017] [Indexed: 12/17/2022]
Abstract
Most metastasizing tumor cells reach distant sites by entering the circulatory system. Within the bloodstream, they are exposed to severe stress due to loss of adhesion to extracellular matrix, hemodynamic shear forces, and attacks of the immune system, and only a few cells manage to extravasate and to form metastases. We review the current understanding of the cellular and molecular mechanisms that allow tumor cells to survive in the intravascular environment and that mediate and promote tumor cell extravasation. As these processes are critical for the metastatic spread of tumor cells, we discuss implications for potential therapeutic approaches and future research.
Collapse
Affiliation(s)
- Boris Strilic
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Stefan Offermanns
- Max Planck Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany; J.W. Goethe University Frankfurt, Center for Molecular Medicine, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| |
Collapse
|
89
|
Blaha L, Zhang C, Cabodi M, Wong JY. A microfluidic platform for modeling metastatic cancer cell matrix invasion. Biofabrication 2017; 9:045001. [PMID: 28812983 DOI: 10.1088/1758-5090/aa869d] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Invasion of the extracellular matrix is a critical step in the colonization of metastatic tumors. The invasion process is thought to be driven by both chemokine signaling and interactions between invading cancer cells and physical components of the metastatic niche, including endothelial cells that line capillary walls and serve as a barrier to both diffusion and invasion of the underlying tissue. Transwell chambers, a tool for generating artificial chemokine gradients to induce cell migration, have facilitated recent work to investigate the chemokine contributions to matrix invasion. These chambers, however, are poorly designed for imaging, which limits their use in investigating the physical cell-cell and cell-matrix interactions driving matrix invasion. Microfluidic devices offer a promising model in which the invasion process can be imaged. Many current designs, however, have limited surface areas and possess intricate geometries that preclude the use of standard staining protocols to visualize cells and matrix proteins. In this work, we present a novel microfluidic platform for imaging cell-cell and cell-matrix interactions driving metastatic cancer cell matrix invasion. Our model is applied to investigate how endothelial cell-secreted matrix proteins and the physical endothelial monolayer itself interact with invading metastatic breast cancer cells to facilitate invasion of an underlying type I collagen gel. The results show that matrix invasion of metastatic breast cancer cells is significantly enhanced in the presence of live endothelial cells. Probing this interaction further, our platform revealed that, while the fibronectin-rich matrix deposited by endothelial cells was not sufficient to drive invasion alone, metastatic breast cancer cells were able to exploit components of energetically inactivated endothelial cells to gain entry into the underlying matrix. These findings reveal novel cell-cell interactions driving a key step in the colonization of metastatic tumors and have important implications for designing drugs targeted at preventing cancer metastasis.
Collapse
Affiliation(s)
- Laura Blaha
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, United States of America
| | | | | | | |
Collapse
|
90
|
Paterson EK, Courtneidge SA. Invadosomes are coming: new insights into function and disease relevance. FEBS J 2017; 285:8-27. [PMID: 28548369 DOI: 10.1111/febs.14123] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/09/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022]
Abstract
Invadopodia and podosomes are discrete, actin-based molecular protrusions that form in cancer cells and normal cells, respectively, in response to diverse signaling pathways and extracellular matrix cues. Although they participate in a host of different cellular processes, they share a common functional theme of controlling pericellular proteolytic activity, which sets them apart from other structures that function in migration and adhesion, including focal adhesions, lamellipodia, and filopodia. In this review, we highlight research that explores the function of these complex structures, including roles for podosomes in embryonic and postnatal development, in angiogenesis and remodeling of the vasculature, in maturation of the postsynaptic membrane, in antigen sampling and recognition, and in cell-cell fusion mechanisms, as well as the involvement of invadopodia at multiple steps of the metastatic cascade, and how all of this may apply in the treatment of human disease states. Finally, we explore recent research that implicates a novel role for exosomes and microvesicles in invadopodia-dependent and invadopodia-independent mechanisms of invasion, respectively.
Collapse
Affiliation(s)
- Elyse K Paterson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Sara A Courtneidge
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
91
|
Jafri MA, Al-Qahtani MH, Shay JW. Role of miRNAs in human cancer metastasis: Implications for therapeutic intervention. Semin Cancer Biol 2017; 44:117-131. [PMID: 28188828 DOI: 10.1016/j.semcancer.2017.02.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/03/2017] [Accepted: 02/06/2017] [Indexed: 12/23/2022]
Abstract
Metastasis is the spread and growth of localized cancer to new locations in the body and is considered the main cause of cancer-related deaths. Metastatic cancer cells display distinct genomic and epigenomic profiles and almost universally an aggressive pathophysiology. A better understanding of the molecular mechanisms and regulation of metastasis, including how metastatic tumors grow and survive in the nascent niche and the interactions of the emergent metastatic cancer cells within the local microenvironment may provide tools to design strategies to restrict metastatic dissemination. Aberrant microRNAs (miRNA) expression has been reported in metastatic cancer cells. MicroRNAs are known to regulate divergent and/or convergent metastatic gene pathways including activation of reprogramming switches during metastasis. An in-depth understanding of role of miRNAs in the metastatic cascade may lead to the identification of novel targets for anti-metastatic therapeutics as well as potential candidate miRNAs for cancer treatment. This review primarily focuses on the role of miRNAs in the mechanisms of cancer metastasis as well as implications for metastatic cancer treatment.
Collapse
Affiliation(s)
- Mohammad Alam Jafri
- Center of Excellence for Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | | | - Jerry William Shay
- Center of Excellence for Genomic Medicine Research, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Cell Biology, University of Texas, Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
92
|
Pulido C, Vendrell I, Ferreira AR, Casimiro S, Mansinho A, Alho I, Costa L. Bone metastasis risk factors in breast cancer. Ecancermedicalscience 2017; 11:715. [PMID: 28194227 PMCID: PMC5295847 DOI: 10.3332/ecancer.2017.715] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Indexed: 12/25/2022] Open
Abstract
Bone is the single most frequent site for bone metastasis in breast cancer patients. Patients with bone-only metastasis have a fairly good prognosis when compared with patients with visceral disease. Nevertheless, cancer-induced bone disease carries an important risk of developing skeletal related events that impact quality of life (QoL). It is therefore particularly important to stratify patients according to their risk of developing bone metastasis. In this context, several risk factors have been studied, including demographic, clinicopathological, genetic, and metabolic factors. Most of them show conflicting or non-definitive associations and are not validated for clinical use. Nonetheless, tumour intrinsic subtype is widely accepted as a major risk factor for bone metastasis development and luminal breast cancer carries an increased risk for bone disease. Other factors such as gene signatures, expression of specific cytokines (such as bone sialoprotein and bone morphogenetic protein 7) or components of the extracellular matrix (like bone crosslinked C-telopeptide) might also influence the development of bone metastasis. Knowledge of risk factors related with bone disease is of paramount importance as it might be a prediction tool for triggering the use of targeted agents and allow for better patient selection for future clinical trials.
Collapse
Affiliation(s)
- Catarina Pulido
- Serviço de Oncologia Médica, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Avenida Professor Egas Moniz, 1649-035 Lisboa, Portugal; These authors contributed equally to this work
| | - Inês Vendrell
- Serviço de Oncologia Médica, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Avenida Professor Egas Moniz, 1649-035 Lisboa, Portugal; These authors contributed equally to this work
| | - Arlindo R Ferreira
- Serviço de Oncologia Médica, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Avenida Professor Egas Moniz, 1649-035 Lisboa, Portugal; Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Sandra Casimiro
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - André Mansinho
- Serviço de Oncologia Médica, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Avenida Professor Egas Moniz, 1649-035 Lisboa, Portugal
| | - Irina Alho
- Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Luís Costa
- Serviço de Oncologia Médica, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Avenida Professor Egas Moniz, 1649-035 Lisboa, Portugal; Luis Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal
| |
Collapse
|
93
|
Cai YD, Zhang Q, Zhang YH, Chen L, Huang T. Identification of Genes Associated with Breast Cancer Metastasis to Bone on a Protein–Protein Interaction Network with a Shortest Path Algorithm. J Proteome Res 2017; 16:1027-1038. [DOI: 10.1021/acs.jproteome.6b00950] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yu-Dong Cai
- School
of Life Sciences, Shanghai University, Shanghai 200444 People’s Republic of China
| | - Qing Zhang
- School
of Life Sciences, Shanghai University, Shanghai 200444 People’s Republic of China
| | - Yu-Hang Zhang
- Institute
of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China
| | - Lei Chen
- College
of Information Engineering, Shanghai Maritime University, Shanghai 201306, People’s Republic of China
| | - Tao Huang
- Institute
of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People’s Republic of China
| |
Collapse
|
94
|
Charles-Orszag A, Lemichez E, Tran Van Nhieu G, Duménil G. Microbial pathogenesis meets biomechanics. Curr Opin Cell Biol 2016; 38:31-7. [PMID: 26849533 DOI: 10.1016/j.ceb.2016.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/22/2015] [Accepted: 01/11/2016] [Indexed: 01/13/2023]
Abstract
Introducing concepts from soft matter physics and mechanics has largely contributed to our understanding of a variety of biological processes. In this review, we argue that this holds true for bacterial pathogenesis. We base this argument on three examples of bacterial pathogens and their interaction with host cells during infection: (i) Shigella flexneri exploits actin-dependent forces to come into close contact with epithelial cells prior to invasion of the epithelium; (ii) Neisseria meningitidis manipulates endothelial cells to resist shear stress during vascular colonization; (iii) bacterial toxins take advantage of the biophysical properties of the host cell plasma membrane to generate transcellular macroapertures in the vascular wall. Together, these examples show that a multidisciplinary approach integrating physics and biology is more necessary than ever to understand complex infectious phenomena. Moreover, this avenue of research will allow the exploration of general processes in cell biology, highlighted by pathogens, in the context of other non-communicable human diseases.
Collapse
Affiliation(s)
- Arthur Charles-Orszag
- Pathogenesis of vascular infections unit, INSERM, Institut Pasteur, 75015 Paris, France
| | - Emmanuel Lemichez
- INSERM, U1065, Microbial Toxins in Host-Pathogen Interactions, Centre Méditerranéen De Médecine Moléculaire, C3M, 151 Route St Antoine de Ginestière, 06204 Nice, France
| | - Guy Tran Van Nhieu
- Equipe Communication Intercellulaire et Infections Microbiennes, Centre de Recherche Interdisciplinaire en Biologie (CIRB), Collège de France, Paris, France; Institut National de la Santé et de la Recherche Médicale U1050, Paris, France; Centre National de la Recherche Scientifique UMR 7241, Paris, France; MEMOLIFE Laboratory of Excellence and Paris Science Lettre, Paris, France
| | - Guillaume Duménil
- Pathogenesis of vascular infections unit, INSERM, Institut Pasteur, 75015 Paris, France.
| |
Collapse
|