1
|
Michael C, Mendonça-Gomes JM, DePaolo CW, Di Cristofano A, de Oliveira S. A zebrafish xenotransplant model of anaplastic thyroid cancer to study tumor microenvironment and innate immune cell interactions in vivo. Endocr Relat Cancer 2024; 31:e230195. [PMID: 38657656 PMCID: PMC11160356 DOI: 10.1530/erc-23-0195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 04/23/2024] [Indexed: 04/26/2024]
Abstract
Anaplastic thyroid cancer (ATC) is of the most aggressive thyroid cancer. While ATC is rare, it accounts for a disproportionately high number of thyroid cancer-related deaths. Here, we developed an ATC xenotransplant model in zebrafish larvae, where we can study tumorigenesis and therapeutic response in vivo. Using both mouse (T4888M) and human (C643)-derived fluorescently labeled ATC cell lines, we show these cell lines display different engraftment rates, mass volume, proliferation, cell death, angiogenic potential, and neutrophil and macrophage recruitment and infiltration. Next, using a PIP-FUCCI reporter to track proliferation in vivo, we observed cells in each phase of the cell cycle. Additionally, we performed long-term non-invasive intravital microscopy over 48 h to understand cellular dynamics in the tumor microenvironment at the single-cell level. Lastly, we tested two drug treatments, AZD2014 and a combination therapy of dabrafenib and trametinib, to show our model could be used as an effective screening platform for new therapeutic compounds for ATC. Altogether, we show that zebrafish xenotransplants make a great model to study thyroid carcinogenesis and the tumor microenvironment, while also being a suitable model to test new therapeutics in vivo.
Collapse
Affiliation(s)
- Cassia Michael
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | | | - Clinton Walton DePaolo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Antonio Di Cristofano
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Cancer Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Cancer Dormancy Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sofia de Oliveira
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine (Hepatology), Albert Einstein College of Medicine, Bronx, NY, USA
- Marion Bessin Liver Research Center, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
- Montefiore-Einstein Cancer Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Cancer Dormancy Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Bian F, Goda C, Wang G, Lan YW, Deng Z, Gao W, Acharya A, Reza AA, Gomez-Arroyo J, Merjaneh N, Ren X, Goveia J, Carmeliet P, Kalinichenko VV, Kalin TV. FOXF1 promotes tumor vessel normalization and prevents lung cancer progression through FZD4. EMBO Mol Med 2024; 16:1063-1090. [PMID: 38589650 PMCID: PMC11099127 DOI: 10.1038/s44321-024-00064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024] Open
Abstract
Cancer cells re-program normal lung endothelial cells (EC) into tumor-associated endothelial cells (TEC) that form leaky vessels supporting carcinogenesis. Transcriptional regulators that control the reprogramming of EC into TEC are poorly understood. We identified Forkhead box F1 (FOXF1) as a critical regulator of EC-to-TEC transition. FOXF1 was highly expressed in normal lung vasculature but was decreased in TEC within non-small cell lung cancers (NSCLC). Low FOXF1 correlated with poor overall survival of NSCLC patients. In mice, endothelial-specific deletion of FOXF1 decreased pericyte coverage, increased vessel permeability and hypoxia, and promoted lung tumor growth and metastasis. Endothelial-specific overexpression of FOXF1 normalized tumor vessels and inhibited the progression of lung cancer. FOXF1 deficiency decreased Wnt/β-catenin signaling in TECs through direct transcriptional activation of Fzd4. Restoring FZD4 expression in FOXF1-deficient TECs through endothelial-specific nanoparticle delivery of Fzd4 cDNA rescued Wnt/β-catenin signaling in TECs, normalized tumor vessels and inhibited the progression of lung cancer. Altogether, FOXF1 increases tumor vessel stability, and inhibits lung cancer progression by stimulating FZD4/Wnt/β-catenin signaling in TECs. Nanoparticle delivery of FZD4 cDNA has promise for future therapies in NSCLC.
Collapse
Affiliation(s)
- Fenghua Bian
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Chinmayee Goda
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Guolun Wang
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Ying-Wei Lan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Zicheng Deng
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Wen Gao
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Anusha Acharya
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Abid A Reza
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Jose Gomez-Arroyo
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Nawal Merjaneh
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA
| | - Xiaomeng Ren
- Division of Asthma Research of Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Jermaine Goveia
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, 3000, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Vladimir V Kalinichenko
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
- Division of Neonatology, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA
| | - Tanya V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA.
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA.
- Department of Internal Medicine, Division of Pulmonary and Critical Care, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA.
| |
Collapse
|
3
|
The inhibition of protein translation promotes tumor angiogenic switch. MOLECULAR BIOMEDICINE 2022; 3:18. [PMID: 35695994 PMCID: PMC9192909 DOI: 10.1186/s43556-022-00081-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
The ‘angiogenic switch’ is critical for tumor progression. However, the pathological details and molecular mechanisms remain incompletely characterized. In this study, we established mammal xenografts in zebrafish to visually investigate the first vessel growth (angiogenic switch) in real-time, by inoculating tumor cells into the perivitelline space of live optically transparent Transgenic (flk1:EGFP) zebrafish larvae. Using this model, we found that hypoxia and hypoxia-inducible factor (HIF) signaling were unnecessary for the angiogenic switch, whereas vascular endothelial growth factor A gene (Vegfa) played a crucial role. Mechanistically, transcriptome analysis showed that the angiogenic switch was characterized by inhibition of translation, but not hypoxia. Phosphorylation of eukaryotic translation initiation factor 2 alpha (Eif2α) and the expression of Vegfa were increased in the angiogenic switch microtumors, and 3D tumor spheroids, and puromycin-treated tumor cells. Vegfa overexpression promoted early onset of the angiogenic switch, whereas Vegfa knockout prevented the first tumor vessel from sprouting. Pretreatment of tumor cells with puromycin promoted the angiogenic switch in vivo similarly to Vegfa overexpression, whereas Vegfa knockdown suppressed the increase. This study provides direc and dynamic in vivo evidences that inhibition of translation, but not hypoxia or HIF signaling promotes the angiogenic switch in tumor by increasing Vegfa transcription.
Collapse
|
4
|
Terrassoux L, Claux H, Bacari S, Meignan S, Furlan A. A Bloody Conspiracy. Blood Vessels and Immune Cells in the Tumor Microenvironment. Cancers (Basel) 2022; 14:cancers14194581. [PMID: 36230504 PMCID: PMC9558972 DOI: 10.3390/cancers14194581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary The tumor microenvironment has risen over the last years as a significant contributor to the failure of antitumoral strategies due to its numerous pro-tumorigenic activities. In this review, we focused on two features of this microenvironment, namely angiogenesis and immunity, which have been the targets of therapies to tackle tumors via its microenvironmental part over the last decade. Increasing our knowledge of the complex interactions within this ecosystem is mandatory to optimize these therapeutic approaches. The development of innovative experimental models is of great help in reaching this goal. Abstract Cancer progression occurs in concomitance with a profound remodeling of the cellular microenvironment. Far from being a mere passive event, the re-orchestration of interactions between the various cell types surrounding tumors highly contributes to the progression of the latter. Tumors notably recruit and stimulate the sprouting of new blood vessels through a process called neo-angiogenesis. Beyond helping the tumor cope with an increased metabolic demand associated with rapid growth, this also controls the metastatic dissemination of cancer cells and the infiltration of immune cells in the tumor microenvironment. To decipher this critical interplay for the clinical progression of tumors, the research community has developed several valuable models in the last decades. This review offers an overview of the various instrumental solutions currently available, including microfluidic chips, co-culture models, and the recent rise of organoids. We highlight the advantages of each technique and the specific questions they can address to better understand the tumor immuno-angiogenic ecosystem. Finally, we discuss this development field’s fundamental and applied perspectives.
Collapse
Affiliation(s)
- Lisa Terrassoux
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Hugo Claux
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Salimata Bacari
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Samuel Meignan
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
| | - Alessandro Furlan
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France
- Tumorigenesis and Resistance to Treatment Unit, Centre Oscar Lambret, F-59000 Lille, France
- Correspondence:
| |
Collapse
|
5
|
Venkatakrishnan G, Parvathi VD. Decoding the mechanism of vascular morphogenesis to explore future prospects in targeted tumor therapy. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:178. [PMID: 36036322 DOI: 10.1007/s12032-022-01810-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/26/2022] [Indexed: 11/27/2022]
Abstract
The growth and formation of blood vessels is an undeniably fundamental biological process crucial to controlling overall development of an organism. This phenomenon consists of two separate processes, commencing with vasculogenesis, which refers to the process of blood vessel formation strictly in embryonic stages, via de novo endothelial cell synthesis. Angiogenesis continues the formation of the vascular network via sprouting and splitting. Tumor growth is dependent on the growth and supply of blood vessels around the tumor mass. Extracellular matrix (ECM) molecules can promote angiogenesis by establishing a vascular network and sequestering pro-angiogenic growth factors. Although the methods by which tumor-associated fibroblasts (which differ in phenotype from normal fibroblasts) influence angiogenesis are unknown, they are thought to be a major source of growth factors and cytokines that attract endothelial cells. Chemokines and growth factors (sourced from macrophages and neutrophils) are also regulators of angiogenesis. When considered as a whole, the tumor microenvironment is a heterogenous and dynamic mass of tissue, composed of a plethora of cell types and an ECM that can fundamentally control the pathological angiogenic switch. Angiogenesis is involved in numerous diseases, and understanding the various mechanisms surrounding this phenomenon is key to finding cures.
Collapse
Affiliation(s)
- Gayathri Venkatakrishnan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, 600116, India
| | - Venkatachalam Deepa Parvathi
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, 600116, India.
| |
Collapse
|
6
|
Abstract
Secretory proteins in tumor tissues are important components of the tumor microenvironment. Secretory proteins act on tumor cells or stromal cells or mediate interactions between tumor cells and stromal cells, thereby affecting tumor progression and clinical treatment efficacy. In this paper, recent research advances in secretory proteins in malignant tumors are reviewed.
Collapse
Affiliation(s)
- Na Zhang
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiajie Hao
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yan Cai
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Mingrong Wang
- State Key Laboratory of Molecular Oncology, Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
7
|
Targen S, Kaya T, Avci ME, Gunes D, Keskus AG, Konu O. ZenoFishDb v1.1: A Database for Xenotransplantation Studies in Zebrafish. Zebrafish 2020; 17:305-318. [PMID: 32931381 DOI: 10.1089/zeb.2020.1869] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Rapidly accumulating literature has proven feasibility of the zebrafish xenograft models in cancer research. Nevertheless, online databases for searching the current zebrafish xenograft literature are in great demand. Herein, we have developed a manually curated database, called ZenoFishDb v1.1 (https://konulab.shinyapps.io/zenofishdb), based on R Shiny platform aiming to provide searchable information on ever increasing collection of zebrafish studies for cancer cell line transplantation and patient-derived xenografts (PDXs). ZenoFishDb v1.1 user interface contains four modules: DataTable, Visualization, PDX Details, and PDX Charts. The DataTable and Visualization pages represent xenograft study details, including injected cell lines, PDX injections, molecular modifications of cell lines, zebrafish strains, as well as technical aspects of the xenotransplantation procedures in table, bar, and/or pie chart formats. The PDX Details module provides comprehensive information on the patient details in table format and can be searched and visualized. Overall, ZenoFishDb v1.1 enables researchers to effectively search, list, and visualize different technical and biological attributes of zebrafish xenotransplantation studies particularly focusing on the new trends that make use of reporters, RNA interference, overexpression, or mutant gene constructs of transplanted cancer cells, stem cells, and PDXs, as well as distinguished host modifications.
Collapse
Affiliation(s)
- Seniye Targen
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Tuğberk Kaya
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey.,Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - M Ender Avci
- Izmir Biomedicine and Genome Center, Dokuz Eylul University, Izmir, Turkey
| | - Damla Gunes
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey
| | - Ayse Gokce Keskus
- Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey
| | - Ozlen Konu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey.,Interdisciplinary Program in Neuroscience, Bilkent University, Ankara, Turkey.,UNAM-Institute of Materials Science and Nanotechnology, Bilkent University, Ankara, Turkey
| |
Collapse
|
8
|
Frantz WT, Ceol CJ. From Tank to Treatment: Modeling Melanoma in Zebrafish. Cells 2020; 9:cells9051289. [PMID: 32455885 PMCID: PMC7290816 DOI: 10.3390/cells9051289] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 12/24/2022] Open
Abstract
Melanoma is the deadliest form of skin cancer and one of few cancers with a growing incidence. A thorough understanding of its pathogenesis is fundamental to developing new strategies to combat mortality and morbidity. Zebrafish—due in large part to their tractable genetics, conserved pathways, and optical properties—have emerged as an excellent system to model melanoma. Zebrafish have been used to study melanoma from a single tumor initiating cell, through metastasis, remission, and finally into relapse. In this review, we examine seminal zebrafish studies that have advanced our understanding of melanoma.
Collapse
Affiliation(s)
- William Tyler Frantz
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA;
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Craig J Ceol
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA;
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Correspondence:
| |
Collapse
|
9
|
Britto DD, Wyroba B, Chen W, Lockwood RA, Tran KB, Shepherd PR, Hall CJ, Crosier KE, Crosier PS, Astin JW. Macrophages enhance Vegfa-driven angiogenesis in an embryonic zebrafish tumour xenograft model. Dis Model Mech 2018; 11:dmm.035998. [PMID: 30396905 PMCID: PMC6307908 DOI: 10.1242/dmm.035998] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/25/2018] [Indexed: 12/11/2022] Open
Abstract
Tumour angiogenesis has long been a focus of anti-cancer therapy; however, anti-angiogenic cancer treatment strategies have had limited clinical success. Tumour-associated myeloid cells are believed to play a role in the resistance of cancer towards anti-angiogenesis therapy, but the mechanisms by which they do this are unclear. An embryonic zebrafish xenograft model has been developed to investigate the mechanisms of tumour angiogenesis and as an assay to screen anti-angiogenic compounds. In this study, we used cell ablation techniques to remove either macrophages or neutrophils and assessed their contribution towards zebrafish xenograft angiogenesis by quantitating levels of graft vascularisation. The ablation of macrophages, but not neutrophils, caused a strong reduction in tumour xenograft vascularisation and time-lapse imaging demonstrated that tumour xenograft macrophages directly associated with the migrating tip of developing tumour blood vessels. Finally, we found that, although macrophages are required for vascularisation in xenografts that either secrete VEGFA or overexpress zebrafish vegfaa, they are not required for the vascularisation of grafts with low levels of VEGFA, suggesting that zebrafish macrophages can enhance Vegfa-driven tumour angiogenesis. The importance of macrophages to this angiogenic response suggests that this model could be used to further investigate the interplay between myeloid cells and tumour vascularisation. Summary: Zebrafish embryonic macrophages associate with the distal tips of tumour xenograft blood vessels and are required for Vegfa-driven angiogenesis.
Collapse
Affiliation(s)
- Denver D Britto
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Barbara Wyroba
- Department of Cell Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków 30-387, Poland
| | - Wenxuan Chen
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Rhoswen A Lockwood
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Khanh B Tran
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Christopher J Hall
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Kathryn E Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Philip S Crosier
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Jonathan W Astin
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland 1023, New Zealand
| |
Collapse
|
10
|
Furlan A, Vercamer C, Heliot L, Wernert N, Desbiens X, Pourtier A. Ets-1 drives breast cancer cell angiogenic potential and interactions between breast cancer and endothelial cells. Int J Oncol 2018; 54:29-40. [PMID: 30365153 PMCID: PMC6254994 DOI: 10.3892/ijo.2018.4605] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 09/10/2018] [Indexed: 12/12/2022] Open
Abstract
Ets-1 transcription factor overexpression in breast cancers is associated with invasive features and is associated with a poor prognosis. Beyond its role in driving carcinoma cell invasion, in this study, we wished to determine whether Ets-1 overexpression in cancer cells promotes angiogenesis by creating a paracrine pro-invasive environment for endothelial cells as well. To address this question, we set up different co-culture models of cancer cells with endothelial cells. Conditioned media from cancer cells induced endothelial cell proliferation, migration and morphogenesis in matrix models. Of note, co-culture assays in three-dimensional matrix models also revealed the reciprocal induction of cancer cell morphogenesis by endothelial cells, in support of an angiocrine action on tumor cells. Ets-1 emerged as a key regulator of the angiogenic potential of breast cancer cells, favoring their ability to induce, in a paracrine manner, the morphogenesis of endothelial cells and also to physically interact with the latter. Nevertheless, Ets-1 overexpression in cancer cells also restrained their chemoattractive potential for endothelial cells both in Boyden chambers and in ex vivo 3D co-cultures. Finally, Ets-1 modulation in breast cancer cells qualitatively altered the angiogenic pattern of experimental in vivo tumors, with a balance between vessel recruitment and intratumoral small capillaries sprouting. Taken together, our data highlight a critical and intriguing role for Ets-1 in the angiogenic potential of breast cancer cells, and reveal another facet of Ets-1 oncogenic activities.
Collapse
Affiliation(s)
- Alessandro Furlan
- UMR8161 CNRS/University of Lille/Pasteur Institute of Lille, Biology Institute of Lille, F-59021 Lille CEDEX, France
| | - Chantal Vercamer
- UMR8161 CNRS/University of Lille/Pasteur Institute of Lille, Biology Institute of Lille, F-59021 Lille CEDEX, France
| | - Laurent Heliot
- UMR8523 PhLAM, CNRS/University of Lille, 59655 Villeneuve d'Ascq CEDEX, France
| | - Nicolas Wernert
- Institute of Pathology, University of Bonn, 53011 Bonn, Germany
| | - Xavier Desbiens
- UMR8161 CNRS/University of Lille/Pasteur Institute of Lille, Biology Institute of Lille, F-59021 Lille CEDEX, France
| | - Albin Pourtier
- UMR8161 CNRS/University of Lille/Pasteur Institute of Lille, Biology Institute of Lille, F-59021 Lille CEDEX, France
| |
Collapse
|
11
|
Brown HK, Schiavone K, Tazzyman S, Heymann D, Chico TJ. Zebrafish xenograft models of cancer and metastasis for drug discovery. Expert Opin Drug Discov 2017; 12:379-389. [PMID: 28277839 DOI: 10.1080/17460441.2017.1297416] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Patients with metastatic cancer suffer the highest rate of cancer-related death, but existing animal models of metastasis have disadvantages that limit our ability to understand this process. The zebrafish is increasingly used for cancer modelling, particularly xenografting of human cancer cell lines, and drug discovery, and may provide novel scientific and therapeutic insights. However, this model system remains underexploited. Areas covered: The authors discuss the advantages and disadvantages of the zebrafish xenograft model for the study of cancer, metastasis and drug discovery. They summarise previous work investigating the metastatic cascade, such as tumour-induced angiogenesis, intravasation, extravasation, dissemination and homing, invasion at secondary sites, assessing metastatic potential and evaluation of cancer stem cells in zebrafish. Expert opinion: The practical advantages of zebrafish for basic biological study and drug discovery are indisputable. However, their ability to sufficiently reproduce and predict the behaviour of human cancer and metastasis remains unproven. For this to be resolved, novel mechanisms must to be discovered in zebrafish that are subsequently validated in humans, and for therapeutic interventions that modulate cancer favourably in zebrafish to successfully translate to human clinical studies. In the meantime, more work is required to establish the most informative methods in zebrafish.
Collapse
Affiliation(s)
- Hannah K Brown
- a Department of Oncology and Metabolism , The Medical School, University of Sheffield , Sheffield , UK.,b Sarcoma Research Unit, Medical School , INSERM, European Associated Laboratory, University of Sheffield , Sheffield , UK
| | - Kristina Schiavone
- a Department of Oncology and Metabolism , The Medical School, University of Sheffield , Sheffield , UK.,b Sarcoma Research Unit, Medical School , INSERM, European Associated Laboratory, University of Sheffield , Sheffield , UK
| | - Simon Tazzyman
- a Department of Oncology and Metabolism , The Medical School, University of Sheffield , Sheffield , UK.,c The Bateson Centre for Lifecourse Biology , University of Sheffield, Western Bank , Sheffield , UK
| | - Dominique Heymann
- a Department of Oncology and Metabolism , The Medical School, University of Sheffield , Sheffield , UK.,b Sarcoma Research Unit, Medical School , INSERM, European Associated Laboratory, University of Sheffield , Sheffield , UK.,d UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours , Nantes University Hospital , Nantes , France.,e Faculty of Medicine , INSERM, UMR 957, Pathophysiology of Bone Resorption and Therapy of Primary Bone Tumours, Equipe Ligue 2012, University of Nantes , Nantes , France
| | - Timothy Ja Chico
- c The Bateson Centre for Lifecourse Biology , University of Sheffield, Western Bank , Sheffield , UK.,f Department of Infection, Immunity & Cardiovascular Disease , The Medical School, University of Sheffield , Sheffield , UK
| |
Collapse
|
12
|
Hu MH, Ma CY, Wang XM, Ye CD, Zhang GX, Chen L, Wang JG. MicroRNA-126 inhibits tumor proliferation and angiogenesis of hepatocellular carcinoma by down-regulating EGFL7 expression. Oncotarget 2016; 7:66922-66934. [PMID: 27611944 PMCID: PMC5341847 DOI: 10.18632/oncotarget.11877] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/12/2016] [Indexed: 12/25/2022] Open
Abstract
This study aims to explore the effects of microRNA-126 (miR-126) on tumor proliferation and angiogenesis of hepatocellular carcinoma (HCC) by targeting EGFL7. HCC tissues and adjacent normal tissues were obtained from 71 HCC patients. Immunohistochemistry (IHC) was conducted to detect expressions of EGFL7 and VEGF and the micro-vessel density (MVD). HCC cell lines were collected and assigned into the blank, miR-126 mimics, miR-126 inhibitors, miR-126 mimics negative control (NC), miR-126 inhibitors NC, si-EGFL7, and miR-126 inhibitors + si-EGFL7 groups. Expressions of miR-126 and EGFL7 mRNA were detected by qRT-PCR assay. The protein expressions of EGFL7 and VEGF were measured by Western blotting. MTT assay was used to measure the proliferation of HCC cells. Tumor xenograft model in nude mice was utilized to evaluate the influence of miR-126 on tumor growth. HCC tissues had higher miR-126 expression and lower EGFL7 mRNA expression than adjacent normal tissues. Compared with the blank, miR-126 mimic NC, miR-126 inhibitor NC and miR-126 inhibitors + si-EGFL7 groups, the protein expressions of EGFL7 and VEGF and cell proliferation were reduced in the miR-126 mimics and si-EGFL7 groups, while the opposite trend was found in the miR-126 inhibitors group. Compared with the blank and miR-126 inhibitors + siRNA-EGFL7 groups, tumor size, tumor weight, and MVD of transplanted tumors in nude mice were significantly reduced in the miR-126 mimics and siRNA-EGFL7 groups, while the opposite trend was found in the miR-126 inhibitors group. In conclusion, miR-126 could inhibit tumor proliferation and angiogenesis of HCC by down-regulating EGFL7 expression.
Collapse
Affiliation(s)
- Ming-Hua Hu
- 1 Department of Surgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, P.R. China
| | - Chen-Yang Ma
- 1 Department of Surgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, P.R. China
| | - Xiao-Ming Wang
- 1 Department of Surgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, P.R. China
| | - Chen-Dong Ye
- 2 Department of Surgery, The Second Affiliated Hospital, Wannan Medical College, Wuhu 241001, P.R. China
| | - Guang-Xian Zhang
- 1 Department of Surgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, P.R. China
| | - Lin Chen
- 1 Department of Surgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, P.R. China
| | - Jin-Guo Wang
- 1 Department of Surgery, Yijishan Hospital, Wannan Medical College, Wuhu 241001, P.R. China
| |
Collapse
|