1
|
Crouigneau R, Li YF, Auxillos J, Goncalves-Alves E, Marie R, Sandelin A, Pedersen SF. Mimicking and analyzing the tumor microenvironment. CELL REPORTS METHODS 2024; 4:100866. [PMID: 39353424 PMCID: PMC11573787 DOI: 10.1016/j.crmeth.2024.100866] [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: 01/28/2024] [Revised: 07/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024]
Abstract
The tumor microenvironment (TME) is increasingly appreciated to play a decisive role in cancer development and response to therapy in all solid tumors. Hypoxia, acidosis, high interstitial pressure, nutrient-poor conditions, and high cellular heterogeneity of the TME arise from interactions between cancer cells and their environment. These properties, in turn, play key roles in the aggressiveness and therapy resistance of the disease, through complex reciprocal interactions between the cancer cell genotype and phenotype, and the physicochemical and cellular environment. Understanding this complexity requires the combination of sophisticated cancer models and high-resolution analysis tools. Models must allow both control and analysis of cellular and acellular TME properties, and analyses must be able to capture the complexity at high depth and spatial resolution. Here, we review the advantages and limitations of key models and methods in order to guide further TME research and outline future challenges.
Collapse
Affiliation(s)
- Roxane Crouigneau
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Yan-Fang Li
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Jamie Auxillos
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Eliana Goncalves-Alves
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rodolphe Marie
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Albin Sandelin
- Section for Computational and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark; Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
| | - Stine Falsig Pedersen
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Zhan T, Song W, Jing G, Yuan Y, Kang N, Zhang Q. Zebrafish live imaging: a strong weapon in anticancer drug discovery and development. Clin Transl Oncol 2024; 26:1807-1835. [PMID: 38514602 DOI: 10.1007/s12094-024-03406-7] [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: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 03/23/2024]
Abstract
Developing anticancer drugs is a complex and time-consuming process. The inability of current laboratory models to reflect important aspects of the tumor in vivo limits anticancer medication research. Zebrafish is a rapid, semi-automated in vivo screening platform that enables the use of non-invasive imaging methods to monitor morphology, survival, developmental status, response to drugs, locomotion, or other behaviors. Zebrafish models are widely used in drug discovery and development for anticancer drugs, especially in conjunction with live imaging techniques. Herein, we concentrated on the use of zebrafish live imaging in anticancer therapeutic research, including drug screening, efficacy assessment, toxicity assessment, and mechanism studies. Zebrafish live imaging techniques have been used in numerous studies, but this is the first time that these techniques have been comprehensively summarized and compared side by side. Finally, we discuss the hypothesis of Zebrafish Composite Model, which may provide future directions for zebrafish imaging in the field of cancer research.
Collapse
Affiliation(s)
- Tiancheng Zhan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Wanqian Song
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Guo Jing
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China.
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Rd, Jinghai District, Tianjin, 301617, People's Republic of China.
| |
Collapse
|
3
|
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
|
4
|
Yadav R, Mahajan S, Singh H, Mehra NK, Madan J, Doijad N, Singh PK, Guru SK. Emerging In Vitro and In Vivo Models: Hope for the Better Understanding of Cancer Progression and Treatment. Adv Biol (Weinh) 2024; 8:e2300487. [PMID: 38581078 DOI: 10.1002/adbi.202300487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/04/2024] [Indexed: 04/07/2024]
Abstract
Various cancer models have been developed to aid the understanding of the underlying mechanisms of tumor development and evaluate the effectiveness of various anticancer drugs in preclinical studies. These models accurately reproduce the critical stages of tumor initiation and development to mimic the tumor microenvironment better. Using these models for target validation, tumor response evaluation, resistance modeling, and toxicity comprehension can significantly enhance the drug development process. Herein, various in vivo or animal models are presented, typically consisting of several mice and in vitro models ranging in complexity from transwell models to spheroids and CRISPR-Cas9 technologies. While in vitro models have been used for decades and dominate the early stages of drug development, they are still limited primary to simplistic tests based on testing on a single cell type cultivated in Petri dishes. Recent advancements in developing new cancer therapies necessitate the generation of complicated animal models that accurately mimic the tumor's complexity and microenvironment. Mice make effective tumor models as they are affordable, have a short reproductive cycle, exhibit rapid tumor growth, and are simple to manipulate genetically. Human cancer mouse models are crucial to understanding the neoplastic process and basic and clinical research improvements. The following review summarizes different in vitro and in vivo metastasis models, their advantages and disadvantages, and their ability to serve as a model for cancer research.
Collapse
Affiliation(s)
- Rachana Yadav
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Hoshiyar Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Neelesh Kumar Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Nandkumar Doijad
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, 500037, India
| | - Santosh Kumar Guru
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| |
Collapse
|
5
|
Michael C, Di Cristofano A, de Oliveira S. A zebrafish xenotransplant model of anaplastic thyroid cancer to study the tumor microenvironment and innate immune cell interactions in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.541816. [PMID: 37398266 PMCID: PMC10312444 DOI: 10.1101/2023.05.29.541816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Anaplastic thyroid cancer (ATC) is a rare malignant subtype of 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, and angiogenic potential. 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 hours to understand cellular dynamics in the tumor microenvironment at the single cell level. Lastly, we tested a well-known mTOR inhibitor to show our model could be used as an effective screening platform for new therapeutic compounds. 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
| | - 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
|
6
|
Shen Y, Sheng R, Guo R. Application of Zebrafish as a Model for Anti-Cancer Activity Evaluation and Toxicity Testing of Natural Products. Pharmaceuticals (Basel) 2023; 16:827. [PMID: 37375774 DOI: 10.3390/ph16060827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Developing natural product-based anti-cancer drugs/agents is a promising way to overcome the serious side effects and toxicity of traditional chemotherapeutics for cancer treatment. However, rapid assessment of the in vivo anti-cancer activities of natural products is a challenge. Alternatively, zebrafish are useful model organisms and perform well in addressing this challenging issue. Nowadays, a growing number of studies have utilized zebrafish models to evaluate the in vivo activities of natural compounds. Herein, we reviewed the application of zebrafish models for evaluating the anti-cancer activity and toxicity of natural products over the past years, summarized its process and benefits, and provided future outlooks for the development of natural product-based anti-cancer drugs.
Collapse
Affiliation(s)
- Yifan Shen
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Ruilong Sheng
- CQM-Centro de Química da Madeira, Campus da Penteada, Universidade da Madeira, 9000-390 Funchal, Portugal
| | - Ruihua Guo
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
- Shanghai Engineering Research Center of Aquatic-Product Processing & Preservation, Shanghai 201306, China
- Laboratory of Quality and Safety Risk Assessment for Aquatic Products on Storage and Preservation (Shanghai), Ministry of Agriculture, Shanghai 201306, China
| |
Collapse
|
7
|
Lichtenegger A, Baumann B, Yasuno Y. Optical Coherence Tomography Is a Promising Tool for Zebrafish-Based Research-A Review. Bioengineering (Basel) 2022; 10:5. [PMID: 36671577 PMCID: PMC9854701 DOI: 10.3390/bioengineering10010005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
The zebrafish is an established vertebrae model in the field of biomedical research. With its small size, rapid maturation time and semi-transparency at early development stages, it has proven to be an important animal model, especially for high-throughput studies. Three-dimensional, high-resolution, non-destructive and label-free imaging techniques are perfectly suited to investigate these animals over various development stages. Optical coherence tomography (OCT) is an interferometric-based optical imaging technique that has revolutionized the diagnostic possibilities in the field of ophthalmology and has proven to be a powerful tool for many microscopic applications. Recently, OCT found its way into state-of-the-art zebrafish-based research. This review article gives an overview and a discussion of the relevant literature and an outlook for this emerging field.
Collapse
Affiliation(s)
- Antonia Lichtenegger
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
- Computational Optics Group, University of Tsukuba, Tsukuba 305-8573, Japan
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, 1090 Vienna, Austria
| | - Yoshiaki Yasuno
- Computational Optics Group, University of Tsukuba, Tsukuba 305-8573, Japan
| |
Collapse
|
8
|
Lichtenegger A, Tamaoki J, Licandro R, Mori T, Mukherjee P, Bian L, Greutter L, Makita S, Wöhrer A, Matsusaka S, Kobayashi M, Baumann B, Yasuno Y. Longitudinal investigation of a xenograft tumor zebrafish model using polarization-sensitive optical coherence tomography. Sci Rep 2022; 12:15381. [PMID: 36100620 PMCID: PMC9470556 DOI: 10.1038/s41598-022-19483-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/30/2022] [Indexed: 01/19/2023] Open
Abstract
Breast cancer is a leading cause of death in female patients worldwide. Further research is needed to get a deeper insight into the mechanisms involved in the development of this devastating disease and to find new therapy strategies. The zebrafish is an established animal model, especially in the field of oncology, which has shown to be a promising candidate for pre-clinical research and precision-based medicine. To investigate cancer growth in vivo in zebrafish, one approach is to explore xenograft tumor models. In this article, we present the investigation of a juvenile xenograft zebrafish model using a Jones matrix optical coherence tomography (JM-OCT) prototype. Immunosuppressed wild-type fish at 1-month post-fertilization were injected with human breast cancer cells and control animals with phosphate buffered saline in the tail musculature. In a longitudinal study, the scatter, polarization, and vasculature changes over time were investigated and quantified in control versus tumor injected animals. A significant decrease in birefringence and an increase in scattering signal was detected in tumor injected zebrafish in comparison to the control once. This work shows the potential of JM-OCT as a non-invasive, label-free, three-dimensional, high-resolution, and tissue-specific imaging tool in pre-clinical cancer research based on juvenile zebrafish models.
Collapse
Affiliation(s)
- Antonia Lichtenegger
- Computational Optics Group, University of Tsukuba, Tsukuba, Japan.
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria.
| | - Junya Tamaoki
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Roxane Licandro
- Computational Imaging Research Lab, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Laboratory for Computational Neuroimaging, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Tomoko Mori
- Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | | | - Lixuan Bian
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Lisa Greutter
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Shuichi Makita
- Computational Optics Group, University of Tsukuba, Tsukuba, Japan
| | - Adelheid Wöhrer
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Satoshi Matsusaka
- Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Makoto Kobayashi
- Department of Molecular and Developmental Biology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Vienna, Austria
| | - Yoshiaki Yasuno
- Computational Optics Group, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
9
|
Tao J, Wei Z, Cheng Y, Xu M, Li Q, Lee SMY, Ge W, Luo KQ, Wang X, Zheng Y. Apoptosis-Sensing Xenograft Zebrafish Tumor Model for Anticancer Evaluation of Redox-Responsive Cross-Linked Pluronic Micelles. ACS APPLIED MATERIALS & INTERFACES 2022; 14:39775-39786. [PMID: 36006680 DOI: 10.1021/acsami.2c09005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
A suitable animal model for preclinical screening and evaluation in vivo could vastly increase the efficiency and success rate of nanomedicine development. Compared with rodents, the transparency of the zebrafish model offers unique advantages of real-time and high-resolution imaging of the whole body and cellular levels in vivo. In this research, we established an apoptosis-sensing xenograft zebrafish tumor model to evaluate the anti-cancer effects of redox-responsive cross-linked Pluronic polymeric micelles (CPPMs) visually and accurately. First, doxorubicin (Dox)-loaded CPPMs were fabricated and characterized with glutathione (GSH)-responsive drug release. Then, the B16F10 xenograft zebrafish tumor model was established to mimic the tumor microenvironment with angiogenesis and high GSH generation for redox-responsive tumor-targeting evaluation in vivo. The high GSH generation was first verified in the xenograft zebrafish tumor model. Compared with ordinary Pluronic polymeric micelles, Dox CPPMs had a much higher accumulation in zebrafish tumor sites. Finally, the apoptosis-sensing B16F10-C3 xenograft zebrafish tumor model was established for visual, rapid, effective, and noninvasive assessment of anti-cancer effects at the cellular level in vivo. The Dox CPPMs significantly inhibited the proliferation of cancer cells and induced apoptosis in the B16F10-C3 xenograft zebrafish tumor model. Therefore, the redox-responsive cross-linked Pluronic micelles showed effective anti-cancer therapy in the xenograft zebrafish tumor model. This xenograft zebrafish tumor model is available for rapid screening and assessment of anti-cancer effects in preclinical studies.
Collapse
Affiliation(s)
- Jinsong Tao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Institute of Applied Physics and Materials Engineering, University of Macau, Macau 999078, China
| | - Zhengjie Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Yaxin Cheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Meng Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Qiuxia Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Wei Ge
- Faculty of Health Sciences, University of Macau, Macau 999078, China
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Macau 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau 999078, China
| | - Xueqing Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
- Faculty of Health Sciences, University of Macau, Macau 999078, China
- MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau 999078, China
| |
Collapse
|
10
|
Basheer F, Dhar P, Samarasinghe RM. Zebrafish Models of Paediatric Brain Tumours. Int J Mol Sci 2022; 23:9920. [PMID: 36077320 PMCID: PMC9456103 DOI: 10.3390/ijms23179920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Paediatric brain cancer is the second most common childhood cancer and is the leading cause of cancer-related deaths in children. Despite significant advancements in the treatment modalities and improvements in the 5-year survival rate, it leaves long-term therapy-associated side effects in paediatric patients. Addressing these impairments demands further understanding of the molecularity and heterogeneity of these brain tumours, which can be demonstrated using different animal models of paediatric brain cancer. Here we review the use of zebrafish as potential in vivo models for paediatric brain tumour modelling, as well as catalogue the currently available zebrafish models used to study paediatric brain cancer pathophysiology, and discuss key findings, the unique attributes that these models add, current challenges and therapeutic significance.
Collapse
Affiliation(s)
- Faiza Basheer
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| | - Poshmaal Dhar
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| | - Rasika M. Samarasinghe
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia
- Instiute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Geelong, VIC 3220, Australia
| |
Collapse
|
11
|
Russo I, Sartor E, Fagotto L, Colombo A, Tiso N, Alaibac M. The Zebrafish model in dermatology: an update for clinicians. Discov Oncol 2022; 13:48. [PMID: 35713744 PMCID: PMC9206045 DOI: 10.1007/s12672-022-00511-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/08/2022] [Indexed: 11/04/2022] Open
Abstract
Recently, the zebrafish has been established as one of the most important model organisms for medical research. Several studies have proved that there is a high level of similarity between human and zebrafish genomes, which encourages the use of zebrafish as a model for understanding human genetic disorders, including cancer. Interestingly, zebrafish skin shows several similarities to human skin, suggesting that this model organism is particularly suitable for the study of neoplastic and inflammatory skin disorders. This paper appraises the specific characteristics of zebrafish skin and describes the major applications of the zebrafish model in dermatological research.
Collapse
Affiliation(s)
- Irene Russo
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy
| | - Emma Sartor
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy
| | - Laura Fagotto
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy
| | - Anna Colombo
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy
| | - Natascia Tiso
- Department of Biology, University of Padua, Via U. Bassi 58/B, 35131, Padua, Italy
| | - Mauro Alaibac
- Unit of Dermatology, University of Padua, Via Gallucci 4, 35128, Padua, Italy.
| |
Collapse
|
12
|
Preclinical models of epithelial ovarian cancer: practical considerations and challenges for a meaningful application. Cell Mol Life Sci 2022; 79:364. [PMID: 35705879 PMCID: PMC9200670 DOI: 10.1007/s00018-022-04395-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/05/2022] [Accepted: 05/23/2022] [Indexed: 12/14/2022]
Abstract
Despite many improvements in ovarian cancer diagnosis and treatment, until now, conventional chemotherapy and new biological drugs have not been shown to cure the disease, and the overall prognosis remains poor. Over 90% of ovarian malignancies are categorized as epithelial ovarian cancers (EOC), a collection of different types of neoplasms with distinctive disease biology, response to chemotherapy, and outcome. Advances in our understanding of the histopathology and molecular features of EOC subtypes, as well as the cellular origins of these cancers, have given a boost to the development of clinically relevant experimental models. The overall goal of this review is to provide a comprehensive description of the available preclinical investigational approaches aimed at better characterizing disease development and progression and at identifying new therapeutic strategies. Systems discussed comprise monolayer (2D) and three-dimensional (3D) cultures of established and primary cancer cell lines, organoids and patient-derived explants, animal models, including carcinogen-induced, syngeneic, genetically engineered mouse, xenografts, patient-derived xenografts (PDX), humanized PDX, and the zebrafish and the laying hen models. Recent advances in tumour-on-a-chip platforms are also detailed. The critical analysis of strengths and weaknesses of each experimental model will aid in identifying opportunities to optimize their translational value.
Collapse
|
13
|
Antonica F, Aiello G, Soldano A, Abballe L, Miele E, Tiberi L. Modeling Brain Tumors: A Perspective Overview of in vivo and Organoid Models. Front Mol Neurosci 2022; 15:818696. [PMID: 35706426 PMCID: PMC9190727 DOI: 10.3389/fnmol.2022.818696] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/23/2022] [Indexed: 11/17/2022] Open
Abstract
Brain tumors are a large and heterogeneous group of neoplasms that affect the central nervous system and include some of the deadliest cancers. Almost all the conventional and new treatments fail to hinder tumoral growth of the most malignant brain tumors. This is due to multiple factors, such as intra-tumor heterogeneity, the microenvironmental properties of the human brain, and the lack of reliable models to test new therapies. Therefore, creating faithful models for each tumor and discovering tailored treatments pose great challenges in the fight against brain cancer. Over the years, different types of models have been generated, and, in this review, we investigated the advantages and disadvantages of the models currently used.
Collapse
Affiliation(s)
- Francesco Antonica
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Giuseppe Aiello
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessia Soldano
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Luana Abballe
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Evelina Miele
- Department of Pediatric Hematology/Oncology and Cellular and Gene Therapy, Bambino Gesù Children’s Hospital, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Luca Tiberi
- Armenise-Harvard Laboratory of Brain Disorders and Cancer, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- *Correspondence: Luca Tiberi,
| |
Collapse
|
14
|
Hason M, Jovicic J, Vonkova I, Bojic M, Simon-Vermot T, White RM, Bartunek P. Bioluminescent Zebrafish Transplantation Model for Drug Discovery. Front Pharmacol 2022; 13:893655. [PMID: 35559262 PMCID: PMC9086674 DOI: 10.3389/fphar.2022.893655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
In the last decade, zebrafish have accompanied the mouse as a robust animal model for cancer research. The possibility of screening small-molecule inhibitors in a large number of zebrafish embryos makes this model particularly valuable. However, the dynamic visualization of fluorescently labeled tumor cells needs to be complemented by a more sensitive, easy, and rapid mode for evaluating tumor growth in vivo to enable high-throughput screening of clinically relevant drugs. In this study we proposed and validated a pre-clinical screening model for drug discovery by utilizing bioluminescence as our readout for the determination of transplanted cancer cell growth and inhibition in zebrafish embryos. For this purpose, we used NanoLuc luciferase, which ensured rapid cancer cell growth quantification in vivo with high sensitivity and low background when compared to conventional fluorescence measurements. This allowed us large-scale evaluation of in vivo drug responses of 180 kinase inhibitors in zebrafish. Our bioluminescent screening platform could facilitate identification of new small-molecules for targeted cancer therapy as well as for drug repurposing.
Collapse
Affiliation(s)
- Martina Hason
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jovana Jovicic
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Ivana Vonkova
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Milan Bojic
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Theresa Simon-Vermot
- Department of Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Richard M. White
- Department of Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Petr Bartunek
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
- CZ-OPENSCREEN, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
15
|
Lichtenegger A, Mukherjee P, Zhu L, Morishita R, Tomita K, Oida D, Leskovar K, Abd El-Sadek I, Makita S, Kirchberger S, Distel M, Baumann B, Yasuno Y. Non-destructive characterization of adult zebrafish models using Jones matrix optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2022; 13:2202-2223. [PMID: 35519284 PMCID: PMC9045912 DOI: 10.1364/boe.455876] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/03/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
The zebrafish is a valuable vertebrate animal model in pre-clinical cancer research. A Jones matrix optical coherence tomography (JM-OCT) prototype operating at 1310 nm and an intensity-based spectral-domain OCT setup at 840 nm were utilized to investigate adult wildtype and a tumor-developing zebrafish model. Various anatomical features were characterized based on their inherent scattering and polarization signature. A motorized translation stage in combination with the JM-OCT prototype enabled large field-of-view imaging to investigate adult zebrafish in a non-destructive way. The diseased animals exhibited tumor-related abnormalities in the brain and near the eye region. The scatter intensity, the attenuation coefficients and local polarization parameters such as the birefringence and the degree of polarization uniformity were analyzed to quantify differences in tumor versus control regions. The proof-of-concept study in a limited number of animals revealed a significant decrease in birefringence in tumors found in the brain and near the eye compared to control regions. The presented work showed the potential of OCT and JM-OCT as non-destructive, high-resolution, and real-time imaging modalities for pre-clinical research based on zebrafish.
Collapse
Affiliation(s)
- Antonia Lichtenegger
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Pradipta Mukherjee
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Lida Zhu
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Rion Morishita
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Kiriko Tomita
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Daisuke Oida
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | - Konrad Leskovar
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
| | - Ibrahim Abd El-Sadek
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
- Department of Physics, Faculty of Science, Damietta University, Egypt
| | - Shuichi Makita
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| | | | - Martin Distel
- St. Anna Children’s Cancer Research Institute (CCRI), Austria
| | - Bernhard Baumann
- Center for Medical Physics and Biomedical Engineering, Medical University of Vienna, Austria
| | - Yoshiaki Yasuno
- Computational Optics Group, Institute of Applied Physics, University of Tsukuba, Japan
| |
Collapse
|
16
|
Functional Therapeutic Target Validation Using Pediatric Zebrafish Xenograft Models. Cancers (Basel) 2022; 14:cancers14030849. [PMID: 35159116 PMCID: PMC8834194 DOI: 10.3390/cancers14030849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Despite the major progress of precision and personalized oncology, a significant therapeutic benefit is only achieved in cases with directly druggable genetic alterations. This highlights the need for additional methods that reliably predict each individual patient’s response in a clinically meaningful time, e.g., through ex vivo functional drug screen profiling. Moreover, patient-derived xenograft (PDX) models are more predictive than cell culture studies, as they reconstruct cell–cell and cell–extracellular matrix (ECM) interactions and consider the tumor microenvironment, drug metabolism and toxicities. Zebrafish PDXs (zPDX) are nowadays emerging as a fast model allowing for multiple drugs to be tested at the same time in a clinically relevant time window. Here, we show that functional drug response profiling of zPDX from primary material obtained through the INdividualized Therapy FOr Relapsed Malignancies in Childhood (INFORM) pediatric precision oncology pipeline provides additional key information for personalized precision oncology. Abstract The survival rate among children with relapsed tumors remains poor, due to tumor heterogeneity, lack of directly actionable tumor drivers and multidrug resistance. Novel personalized medicine approaches tailored to each tumor are urgently needed to improve cancer treatment. Current pediatric precision oncology platforms, such as the INFORM (INdividualized Therapy FOr Relapsed Malignancies in Childhood) study, reveal that molecular profiling of tumor tissue identifies targets associated with clinical benefit in a subgroup of patients only and should be complemented with functional drug testing. In such an approach, patient-derived tumor cells are exposed to a library of approved oncological drugs in a physiological setting, e.g., in the form of animal avatars injected with patient tumor cells. We used molecularly fully characterized tumor samples from the INFORM study to compare drug screen results of individual patient-derived cell models in functional assays: (i) patient-derived spheroid cultures within a few days after tumor dissociation; (ii) tumor cells reisolated from the corresponding mouse PDX; (iii) corresponding long-term organoid-like cultures and (iv) drug evaluation with the corresponding zebrafish PDX (zPDX) model. Each model had its advantage and complemented the others for drug hit and drug combination selection. Our results provide evidence that in vivo zPDX drug screening is a promising add-on to current functional drug screening in precision medicine platforms.
Collapse
|
17
|
Wang C, Liu S, Tang KFJ, Zhang Q. Natural infection of covert mortality nodavirus affects Zebrafish (Danio rerio). JOURNAL OF FISH DISEASES 2021; 44:1315-1324. [PMID: 34101847 DOI: 10.1111/jfd.13390] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/22/2021] [Accepted: 04/22/2021] [Indexed: 06/12/2023]
Abstract
Covert mortality nodavirus (CMNV), a novel aquatic pathogen, causes viral covert mortality disease (VCMD) in shrimps and also known to infect farmed marine fish. To date, there has no report regarding the ability of this virus to infect freshwater fish. In this study, we screened and discovered CMNV-positive freshwater zebrafish individuals by reverse transcription-nested PCR (RT-nPCR). The sequence of CMNV amplicons from zebrafish was found to share 99% identity with RNA-dependent RNA polymerase (RdRp) gene of the original CMNV isolate. Histopathological examination of the CMNV-positive zebrafish samples revealed extensive vacuolation and karyopyknosis lesions in the retina of the eye and the midbrain mesencephalon. CMNV-like virus particles were visualized in these tissues under transmission electron microscope. Different degrees of pathological damages were also found in muscle, gills, thymus and ovarian tissues. Strong positive signals of CMNV probe were observed in these infected tissues by in situ hybridization. Overall, all results indicated that zebrafish, an acknowledged model organism, could be infected naturally by CMNV. Thus, it is needed to pay close attention to the possible interference of CMNV whether in assessment of toxic substances, or in studying the developmental characterization and the nerval function, when zebrafish was used as model animal.
Collapse
Affiliation(s)
- Chong Wang
- Function Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Shuang Liu
- Function Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Kathy F J Tang
- Function Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Qingli Zhang
- Function Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Key Laboratory of Maricultural Organism Disease Control, Ministry of Agriculture, Qingdao Key Laboratory of Mariculture Epidemiology and Biosecurity, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| |
Collapse
|
18
|
Tucker ER, George S, Angelini P, Bruna A, Chesler L. The Promise of Patient-Derived Preclinical Models to Accelerate the Implementation of Personalised Medicine for Children with Neuroblastoma. J Pers Med 2021; 11:248. [PMID: 33808071 PMCID: PMC8065808 DOI: 10.3390/jpm11040248] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 01/02/2023] Open
Abstract
Patient-derived preclinical models are now a core component of cancer research and have the ability to drastically improve the predictive power of preclinical therapeutic studies. However, their development and maintenance can be challenging, time consuming, and expensive. For neuroblastoma, a developmental malignancy of the neural crest, it is possible to establish patient-derived models as xenografts in mice and zebrafish, and as spheroids and organoids in vitro. These varied approaches have contributed to comprehensive packages of preclinical evidence in support of new therapeutics for neuroblastoma. We discuss here the ethical and technical considerations for the creation of patient-derived models of neuroblastoma and how their use can be optimized for the study of tumour evolution and preclinical therapies. We also discuss how neuroblastoma patient-derived models might become avatars for personalised medicine for children with this devastating disease.
Collapse
Affiliation(s)
- Elizabeth R. Tucker
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Cotswold Road, London SM2 5NG, UK; (E.R.T.); (S.G.)
| | - Sally George
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Cotswold Road, London SM2 5NG, UK; (E.R.T.); (S.G.)
| | - Paola Angelini
- Children and Young People’s Unit, The Royal Marsden, Downs Road, Sutton, Surrey SM2 5PT, UK;
| | - Alejandra Bruna
- Preclinical Paediatric Cancer Evolution, Centre for Cancer Drug Discovery, The Institute of Cancer Research, Cotswold Road, London SM2 5NG, UK;
| | - Louis Chesler
- Paediatric Tumour Biology, Division of Clinical Studies, The Institute of Cancer Research, Cotswold Road, London SM2 5NG, UK; (E.R.T.); (S.G.)
| |
Collapse
|
19
|
Dietrich K, Fiedler IA, Kurzyukova A, López-Delgado AC, McGowan LM, Geurtzen K, Hammond CL, Busse B, Knopf F. Skeletal Biology and Disease Modeling in Zebrafish. J Bone Miner Res 2021; 36:436-458. [PMID: 33484578 DOI: 10.1002/jbmr.4256] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022]
Abstract
Zebrafish are teleosts (bony fish) that share with mammals a common ancestor belonging to the phylum Osteichthyes, from which their endoskeletal systems have been inherited. Indeed, teleosts and mammals have numerous genetically conserved features in terms of skeletal elements, ossification mechanisms, and bone matrix components in common. Yet differences related to bone morphology and function need to be considered when investigating zebrafish in skeletal research. In this review, we focus on zebrafish skeletal architecture with emphasis on the morphology of the vertebral column and associated anatomical structures. We provide an overview of the different ossification types and osseous cells in zebrafish and describe bone matrix composition at the microscopic tissue level with a focus on assessing mineralization. Processes of bone formation also strongly depend on loading in zebrafish, as we elaborate here. Furthermore, we illustrate the high regenerative capacity of zebrafish bones and present some of the technological advantages of using zebrafish as a model. We highlight zebrafish axial and fin skeleton patterning mechanisms, metabolic bone disease such as after immunosuppressive glucocorticoid treatment, as well as osteogenesis imperfecta (OI) and osteopetrosis research in zebrafish. We conclude with a view of why larval zebrafish xenografts are a powerful tool to study bone metastasis. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Kristin Dietrich
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Imke Ak Fiedler
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anastasia Kurzyukova
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Alejandra C López-Delgado
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Lucy M McGowan
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Karina Geurtzen
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| | - Chrissy L Hammond
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Interdisciplinary Competence Center for Interface Research (ICCIR), Hamburg, Germany
| | - Franziska Knopf
- Center for Regenerative Therapies TU Dresden (CRTD), Center for Healthy Aging TU Dresden, Dresden, Germany
| |
Collapse
|
20
|
Cabezas-Sainz P, Coppel C, Pensado-López A, Fernandez P, Muinelo-Romay L, López-López R, Rubiolo JA, Sánchez L. Morphological Abnormalities and Gene Expression Changes Caused by High Incubation Temperatures in Zebrafish Xenografts with Human Cancer Cells. Genes (Basel) 2021; 12:genes12010113. [PMID: 33477746 PMCID: PMC7832305 DOI: 10.3390/genes12010113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/09/2021] [Accepted: 01/13/2021] [Indexed: 11/24/2022] Open
Abstract
Published studies show that most of the human cancer xenograft studies in zebrafish embryos have used incubation temperatures in the range of 32–34 °C for 3–6 days post-injection, trying to find a compromise temperature between the zebrafish embryos (28 °C) and the human injected cells (37 °C). While this experimental setup is widely used, a question remains: is possible to overcome the drawbacks caused by a suboptimal temperature for the injected cells? To clarify the effect of temperature and injected cells on the host, in this study, we analyzed the development and health of the last in response to different temperatures in the presence or absence of injected human cancer cells. Comparing different incubation temperatures (28, 34 and 36 °C), we determined morphological abnormalities and developmental effects in injected and non-injected embryos at different time points. Besides this, the expression of selected genes was determined by qPCR to determine temperature affected metabolic processes in the embryos. The results indicate that an incubation temperature of 36 °C during a period of 48 h is suitable for xenotransplantation without morphological or metabolic changes that could be affecting the host or the injected cells, allowing them to proliferate near their optimal temperature.
Collapse
Affiliation(s)
- Pablo Cabezas-Sainz
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (C.C.); (A.P.-L.); (P.F.)
| | - Carlos Coppel
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (C.C.); (A.P.-L.); (P.F.)
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (C.C.); (A.P.-L.); (P.F.)
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Pedro Fernandez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (C.C.); (A.P.-L.); (P.F.)
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 29029 Madrid, Spain
| | - Rafael López-López
- Translational Laboratory, Medical Oncology Department, Complexo Hospitalario Universitario de Santiago de Compostela/SERGAS, 15706 Santiago de Compostela, Spain;
| | - Juan A. Rubiolo
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (C.C.); (A.P.-L.); (P.F.)
- Correspondence: (J.A.R.); (L.S.); Tel.: +34-982-822-429 (L.S.)
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (C.C.); (A.P.-L.); (P.F.)
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Correspondence: (J.A.R.); (L.S.); Tel.: +34-982-822-429 (L.S.)
| |
Collapse
|
21
|
Studying the Tumor Microenvironment in Zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1329:69-92. [PMID: 34664234 DOI: 10.1007/978-3-030-73119-9_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment significantly contributes to tumor initiation, progression, neo-angiogenesis, and metastasis, and a better understanding of the role of the different cellular players would facilitate the development of novel therapeutic strategies for cancer treatment. Towards this goal, intravital imaging is a powerful method to unravel interaction partners of tumor cells. Among vertebrate model organisms, zebrafish is uniquely suited for in vivo imaging studies. In recent years zebrafish has also become a valuable model in cancer research. In this chapter, we will summarize, how zebrafish has been used to characterize cells of the tumor microenvironment. We will cover both genetically engineered cancer models and xenograft models in zebrafish. The majority of work has been done on the role of innate immune cells and their role during tumor initiation and metastasis, but we will also cover studies focusing on adipocytes, fibroblasts, and endothelial cells. Taken together, we will highlight the versatile use of the zebrafish model for in vivo tumor microenvironment studies.
Collapse
|
22
|
An Integrated In Silico and In Vivo Approach to Identify Protective Effects of Palonosetron in Cisplatin-Induced Nephrotoxicity. Pharmaceuticals (Basel) 2020; 13:ph13120480. [PMID: 33419241 PMCID: PMC7766590 DOI: 10.3390/ph13120480] [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: 11/30/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 11/17/2022] Open
Abstract
Cisplatin is widely used to treat various types of cancers, but it is often limited by nephrotoxicity. Here, we employed an integrated in silico and in vivo approach to identify potential treatments for cisplatin-induced nephrotoxicity (CIN). Using publicly available mouse kidney and human kidney organoid transcriptome datasets, we first identified a 208-gene expression signature for CIN and then used the bioinformatics database Cmap and Lincs Unified Environment (CLUE) to identify drugs expected to counter the expression signature for CIN. We also searched the adverse event database, Food and Drug Administration. Adverse Event Reporting System (FAERS), to identify drugs that reduce the reporting odds ratio of developing cisplatin-induced acute kidney injury. Palonosetron, a serotonin type 3 receptor (5-hydroxytryptamine receptor 3 (5-HT3R)) antagonist, was identified by both CLUE and FAERS analyses. Notably, clinical data from 103 patients treated with cisplatin for head and neck cancer revealed that palonosetron was superior to ramosetron in suppressing cisplatin-induced increases in serum creatinine and blood urea nitrogen levels. Moreover, palonosetron significantly increased the survival rate of zebrafish exposed to cisplatin but not to other 5-HT3R antagonists. These results not only suggest that palonosetron can suppress CIN but also support the use of in silico and in vivo approaches in drug repositioning studies.
Collapse
|
23
|
Rapid In Vivo Validation of HDAC Inhibitor-Based Treatments in Neuroblastoma Zebrafish Xenografts. Pharmaceuticals (Basel) 2020; 13:ph13110345. [PMID: 33121173 PMCID: PMC7692187 DOI: 10.3390/ph13110345] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/23/2020] [Accepted: 10/26/2020] [Indexed: 01/01/2023] Open
Abstract
The survival rate among children with relapsed neuroblastomas continues to be poor, and thus new therapeutic approaches identified by reliable preclinical drug testing models are urgently needed. Zebrafish are a powerful vertebrate model in preclinical cancer research. Here, we describe a zebrafish neuroblastoma yolk sac model to evaluate efficacy and toxicity of histone deacetylase (HDAC) inhibitor treatments. Larvae were engrafted with fluorescently labeled, genetically diverse, established cell lines and short-term cultures of patient-derived primary cells. Engrafted tumors progressed locally and disseminated remotely in an intact environment. Combination treatments involving the standard chemotherapy doxorubicin and HDAC inhibitors substantially reduced tumor volume, induced tumor cell death, and inhibited tumor cell dissemination to the tail region. Hence, this model allows for fast, cost-efficient, and reliable in vivo evaluation of toxicity and response of the primary and metastatic tumor sites to drug combinations.
Collapse
|
24
|
Mary B, Ghoroghi S, Hyenne V, Goetz JG. Live tracking of extracellular vesicles in larval zebrafish. Methods Enzymol 2020; 645:243-275. [PMID: 33565975 DOI: 10.1016/bs.mie.2020.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Formerly considered as insignificant cell debris, extracellular vesicles (EVs) have emerged as potent mediators of cell-cell communication, both in proximity and at distance from the producing cell. EVs are transported in body fluids and can be internalized by specific distant cells to ultimately deliver a functional message. Despite their striking importance in many physiological and pathological contexts, the exact mechanisms by which EVs impose local and distant modifications of the microenvironment in vivo remain to be fully understood. We realized that some conceptual gaps are direct consequences of the difficulty to visualize the shuttling and targeting of EVs in real time in vivo. The zebrafish larvae offered attractive features for live tracking of EVs, within circulating fluids. Here, we describe the experimental procedures that we have built for dissecting the dissemination of EVs at high spatio-temporal resolution in vivo.
Collapse
Affiliation(s)
- Benjamin Mary
- INSERM UMR_S1109, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Shima Ghoroghi
- INSERM UMR_S1109, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Vincent Hyenne
- INSERM UMR_S1109, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France; CNRS, SNC 5055, Strasbourg, France.
| | - Jacky G Goetz
- INSERM UMR_S1109, Strasbourg, France; Université de Strasbourg, Strasbourg, France; Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
| |
Collapse
|
25
|
Cabezas-Sáinz P, Pensado-López A, Sáinz B, Sánchez L. Modeling Cancer Using Zebrafish Xenografts: Drawbacks for Mimicking the Human Microenvironment. Cells 2020; 9:E1978. [PMID: 32867288 PMCID: PMC7564051 DOI: 10.3390/cells9091978] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/07/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023] Open
Abstract
The first steps towards establishing xenografts in zebrafish embryos were performed by Lee et al., 2005 and Haldi et al., 2006, paving the way for studying human cancers using this animal species. Since then, the xenograft technique has been improved in different ways, ranging from optimizing the best temperature for xenografted embryo incubation, testing different sites for injection of human tumor cells, and even developing tools to study how the host interacts with the injected cells. Nonetheless, a standard protocol for performing xenografts has not been adopted across laboratories, and further research on the temperature, microenvironment of the tumor or the cell-host interactions inside of the embryo during xenografting is still needed. As a consequence, current non-uniform conditions could be affecting experimental results in terms of cell proliferation, invasion, or metastasis; or even overestimating the effects of some chemotherapeutic drugs on xenografted cells. In this review, we highlight and raise awareness regarding the different aspects of xenografting that need to be improved in order to mimic, in a more efficient way, the human tumor microenvironment, resulting in more robust and accurate in vivo results.
Collapse
Affiliation(s)
- Pablo Cabezas-Sáinz
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
- Genomic Medicine Group, Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Bruno Sáinz
- Departamento de Bioquímica, Facultad de Medicina, Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, Universidad Autónoma de Madrid, Arzobispo Morcillo 4, 28029 Madrid, Spain;
- Cancer Stem Cell and Fibroinflammatory Microenvironment Group, Chronic Diseases and Cancer Area 3-Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034 Madrid, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (P.C.-S.); (A.P.-L.)
| |
Collapse
|
26
|
Rubbini D, Cornet C, Terriente J, Di Donato V. CRISPR Meets Zebrafish: Accelerating the Discovery of New Therapeutic Targets. SLAS DISCOVERY 2020; 25:552-567. [PMID: 32462967 DOI: 10.1177/2472555220926920] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Bringing a new drug to the market costs an average of US$2.6 billion and takes more than 10 years from discovery to regulatory approval. Despite the need to reduce cost and time to increase productivity, pharma companies tend to crowd their efforts in the same indications and drug targets. This results in the commercialization of drugs that share the same mechanism of action (MoA) and, in many cases, equivalent efficacies among them-an outcome that helps neither patients nor the balance sheet of the companies trying to bring therapeutics to the same patient population. Indeed, the discovery of new therapeutic targets, based on a deeper understanding of the disease biology, would likely provide more innovative MoAs and potentially greater drug efficacies. It would also bring better chances for identifying appropriate treatments according to the patient's genetic stratification. Nowadays, we count with an enormous amount of unprocessed information on potential disease targets that could be extracted from omics data obtained from patient samples. In addition, hundreds of pharmacological and genetic screenings have been performed to identify innovative drug targets. Traditionally, rodents have been the animal models of choice to perform functional genomic studies. The high experimental cost, combined with the low throughput provided by those models, however, is a bottleneck for discovering and validating novel genetic disease associations. To overcome these limitations, we propose that zebrafish, in conjunction with the use of CRISPR/Cas9 genome-editing tools, could streamline functional genomic processes to bring biologically relevant knowledge on innovative disease targets in a shorter time frame.
Collapse
Affiliation(s)
- Davide Rubbini
- ZeClinics SL, IGTP (Germans Trias I Pujol Research Institute), Barcelona, Spain
| | - Carles Cornet
- ZeClinics SL, IGTP (Germans Trias I Pujol Research Institute), Barcelona, Spain
| | - Javier Terriente
- ZeClinics SL, IGTP (Germans Trias I Pujol Research Institute), Barcelona, Spain
| | - Vincenzo Di Donato
- ZeClinics SL, IGTP (Germans Trias I Pujol Research Institute), Barcelona, Spain
| |
Collapse
|
27
|
Dahlem C, Siow WX, Lopatniuk M, Tse WKF, Kessler SM, Kirsch SH, Hoppstädter J, Vollmar AM, Müller R, Luzhetskyy A, Bartel K, Kiemer AK. Thioholgamide A, a New Anti-Proliferative Anti-Tumor Agent, Modulates Macrophage Polarization and Metabolism. Cancers (Basel) 2020; 12:cancers12051288. [PMID: 32438733 PMCID: PMC7281193 DOI: 10.3390/cancers12051288] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/24/2022] Open
Abstract
Natural products represent powerful tools searching for novel anticancer drugs. Thioholgamide A (thioA) is a ribosomally synthesized and post-translationally modified peptide, which has been identified as a product of Streptomyces sp. MUSC 136T. In this study, we provide a comprehensive biological profile of thioA, elucidating its effects on different hallmarks of cancer in tumor cells as well as in macrophages as crucial players of the tumor microenvironment. In 2D and 3D in vitro cell culture models thioA showed potent anti-proliferative activities in cancer cells at nanomolar concentrations. Anti-proliferative actions were confirmed in vivo in zebrafish embryos. Cytotoxicity was only induced at several-fold higher concentrations, as assessed by live-cell microscopy and biochemical analyses. ThioA exhibited a potent modulation of cell metabolism by inhibiting oxidative phosphorylation, as determined in a live-cell metabolic assay platform. The metabolic modulation caused a repolarization of in vitro differentiated and polarized tumor-promoting human monocyte-derived macrophages: ThioA-treated macrophages showed an altered morphology and a modulated expression of genes and surface markers. Taken together, the metabolic regulator thioA revealed low activities in non-tumorigenic cells and an interesting anti-cancer profile by orchestrating different hallmarks of cancer, both in tumor cells as well as in macrophages as part of the tumor microenvironment.
Collapse
Affiliation(s)
- Charlotte Dahlem
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Campus C2 3, 66123 Saarbrücken, Germany; (C.D.); (S.M.K.); (J.H.)
| | - Wei Xiong Siow
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; (W.X.S.); (A.M.V.); (K.B.)
| | - Maria Lopatniuk
- Department of Pharmacy, Pharmaceutical Biotechnology, Saarland University, Campus C2 3, 66123 Saarbrücken, Germany; (M.L.); (A.L.)
| | - William K. F. Tse
- Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, 744 Motooka Nishi-ku, Fukuoka 819-0395, Japan;
| | - Sonja M. Kessler
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Campus C2 3, 66123 Saarbrücken, Germany; (C.D.); (S.M.K.); (J.H.)
- Department of Pharmacology for Natural Sciences, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Susanne H. Kirsch
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Campus E8 1, 66123 Saarbrücken, Germany; (S.H.K.); (R.M.)
| | - Jessica Hoppstädter
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Campus C2 3, 66123 Saarbrücken, Germany; (C.D.); (S.M.K.); (J.H.)
| | - Angelika M. Vollmar
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; (W.X.S.); (A.M.V.); (K.B.)
| | - Rolf Müller
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Campus E8 1, 66123 Saarbrücken, Germany; (S.H.K.); (R.M.)
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Andriy Luzhetskyy
- Department of Pharmacy, Pharmaceutical Biotechnology, Saarland University, Campus C2 3, 66123 Saarbrücken, Germany; (M.L.); (A.L.)
- Department of Microbial Natural Products, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, Campus E8 1, 66123 Saarbrücken, Germany; (S.H.K.); (R.M.)
| | - Karin Bartel
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Butenandtstrasse 5-13, 81377 Munich, Germany; (W.X.S.); (A.M.V.); (K.B.)
| | - Alexandra K. Kiemer
- Department of Pharmacy, Pharmaceutical Biology, Saarland University, Campus C2 3, 66123 Saarbrücken, Germany; (C.D.); (S.M.K.); (J.H.)
- Correspondence: ; Tel.: +49-681-302-57301
| |
Collapse
|
28
|
Fazio M, Ablain J, Chuan Y, Langenau DM, Zon LI. Zebrafish patient avatars in cancer biology and precision cancer therapy. Nat Rev Cancer 2020; 20:263-273. [PMID: 32251397 PMCID: PMC8011456 DOI: 10.1038/s41568-020-0252-3] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/05/2020] [Indexed: 01/05/2023]
Abstract
In precision oncology, two major strategies are being pursued for predicting clinically relevant tumour behaviours, such as treatment response and emergence of drug resistance: inference based on genomic, transcriptomic, epigenomic and/or proteomic analysis of patient samples, and phenotypic assays in personalized cancer avatars. The latter approach has historically relied on in vivo mouse xenografts and in vitro organoids or 2D cell cultures. Recent progress in rapid combinatorial genetic modelling, the development of a genetically immunocompromised strain for xenotransplantation of human patient samples in adult zebrafish and the first clinical trial using xenotransplantation in zebrafish larvae for phenotypic testing of drug response bring this tiny vertebrate to the forefront of the precision medicine arena. In this Review, we discuss advances in transgenic and transplantation-based zebrafish cancer avatars, and how these models compare with and complement mouse xenografts and human organoids. We also outline the unique opportunities that these different models present for prediction studies and current challenges they face for future clinical deployment.
Collapse
Affiliation(s)
- Maurizio Fazio
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Julien Ablain
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Yan Chuan
- Molecular Pathology Unit, Cancer Center, Massachusetts General Hospital Research Institute, Charlestown, MA, USA
| | - David M Langenau
- Molecular Pathology Unit, Cancer Center, Massachusetts General Hospital Research Institute, Charlestown, MA, USA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
29
|
Xiao J, Glasgow E, Agarwal S. Zebrafish Xenografts for Drug Discovery and Personalized Medicine. Trends Cancer 2020; 6:569-579. [PMID: 32312681 DOI: 10.1016/j.trecan.2020.03.012] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/18/2020] [Accepted: 03/24/2020] [Indexed: 02/06/2023]
Abstract
Cancer is the second leading cause of death in the world. Given that cancer is a highly individualized disease, predicting the best chemotherapeutic treatment for individual patients can be difficult. Ex vivo models such as mouse patient-derived xenografts (PDX) and organoids are being developed to predict patient-specific chemosensitivity profiles before treatment in the clinic. Although promising, these models have significant disadvantages including long growth times that introduce genetic and epigenetic changes to the tumor. The zebrafish xenograft assay is ideal for personalized medicine. Imaging of the small, transparent fry is unparalleled among vertebrate organisms. In addition, the speed (5-7 days) and small patient tissue requirements (100-200 cells per animal) are unique features of the zebrafish xenograft model that enable patient-specific chemosensitivity analyses.
Collapse
Affiliation(s)
- Jerry Xiao
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Eric Glasgow
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA.
| | - Seema Agarwal
- Department of Pathology, Center for Cell Reprogramming, Georgetown University Medical Center, Washington, DC 20007, USA.
| |
Collapse
|
30
|
Park G, Burroughs-Garcia J, Foster CA, Hasan A, Borga C, Frazer JK. Zebrafish B cell acute lymphoblastic leukemia: new findings in an old model. Oncotarget 2020; 11:1292-1305. [PMID: 32341750 PMCID: PMC7170496 DOI: 10.18632/oncotarget.27555] [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: 11/13/2019] [Accepted: 03/19/2020] [Indexed: 12/22/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric, and ninth most common adult, cancer. ALL can develop in either B or T lymphocytes, but B-lineage ALL (B-ALL) exceeds T-ALL clinically. As for other cancers, animal models allow study of the molecular mechanisms driving ALL. Several zebrafish (Danio rerio) T-ALL models have been reported, but until recently, robust D. rerio B-ALL models were not described. Then, D. rerio B-ALL was discovered in two related zebrafish transgenic lines; both were already known to develop T-ALL. Here, we report new B-ALL findings in one of these models, fish expressing transgenic human MYC (hMYC). We describe B-ALL incidence in a large cohort of hMYC fish, and show B-ALL in two new lines where T-ALL does not interfere with B-ALL detection. We also demonstrate B-ALL responses to steroid and radiation treatments, which effect ALL remissions, but are usually followed by prompt relapses. Finally, we report gene expression in zebrafish B lymphocytes and B-ALL, in both bulk samples and single B- and T-ALL cells. Using these gene expression profiles, we compare differences between the two new D. rerio B-ALL models, which are both driven by transgenic mammalian MYC oncoproteins. Collectively, these new data expand the utility of this new vertebrate B-ALL model.
Collapse
Affiliation(s)
- Gilseung Park
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,These authors contributed equally to this work
| | - Jessica Burroughs-Garcia
- Department of Pediatrics, Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,These authors contributed equally to this work
| | - Clay A Foster
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA.,These authors contributed equally to this work
| | - Ameera Hasan
- Department of Microbiology & Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Chiara Borga
- Department of Pediatrics, Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - J Kimble Frazer
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,Department of Pediatrics, Section of Pediatric Hematology-Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,Department of Microbiology & Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
31
|
Zebrafish Avatars towards Personalized Medicine-A Comparative Review between Avatar Models. Cells 2020; 9:cells9020293. [PMID: 31991800 PMCID: PMC7072137 DOI: 10.3390/cells9020293] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/08/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer frequency and prevalence have been increasing in the past decades, with devastating impacts on patients and their families. Despite the great advances in targeted approaches, there is still a lack of methods to predict individual patient responses, and therefore treatments are tailored according to average response rates. “Omics” approaches are used for patient stratification and choice of therapeutic options towards a more precise medicine. These methods, however, do not consider all genetic and non-genetic dynamic interactions that occur upon drug treatment. Therefore, the need to directly challenge patient cells in a personalized manner remains. The present review addresses the state of the art of patient-derived in vitro and in vivo models, from organoids to mouse and zebrafish Avatars. The predictive power of each model based on the retrospective correlation with the patient clinical outcome will be considered. Finally, the review is focused on the emerging zebrafish Avatars and their unique characteristics allowing a fast analysis of local and systemic effects of drug treatments at the single-cell level. We also address the technical challenges that the field has yet to overcome.
Collapse
|
32
|
Nolan JC, Frawley T, Tighe J, Soh H, Curtin C, Piskareva O. Preclinical models for neuroblastoma: Advances and challenges. Cancer Lett 2020; 474:53-62. [PMID: 31962141 DOI: 10.1016/j.canlet.2020.01.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/12/2022]
Abstract
Neuroblastoma is a paediatric cancer of the sympathetic nervous system and the most common solid tumour of infancy, contributing to 15% of paediatric oncology deaths. Current therapies are not effective in the long-term treatment of almost 80% of patients with this clinically aggressive disease. The primary challenge in the identification and validation of new agents for paediatric drug development is the accurate representation of tumour biology and diversity. In addition to this limitation, the low incidence of neuroblastoma makes the recruitment of eligible patients for early phase clinical trials highly challenging and highlights the need for robust preclinical testing to ensure that the best treatments are selected. The research field requires new preclinical models, technologies, and concepts to tackle these problems. Tissue engineering offers attractive tools to assist in the development of three-dimensional (3D) cell models using various biomaterials and manufacturing approaches that recreate the geometry, mechanics, heterogeneity, metabolic gradients, and cell communication of the native tumour microenvironment. In this review, we discuss current experimental models and assess their abilities to reflect the structural organisation and physiological conditions of the human body, in addition to current and new techniques to recapitulate the tumour niche using tissue-engineered platforms. Finally, we will discuss the possible use of novel 3D in vitro culture systems to address open questions in neuroblastoma biology.
Collapse
Affiliation(s)
- J C Nolan
- Cancer Bio-Engineering Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - T Frawley
- Cancer Bio-Engineering Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| | - J Tighe
- Cancer Bio-Engineering Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - H Soh
- Cancer Bio-Engineering Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - C Curtin
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - O Piskareva
- Cancer Bio-Engineering Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland; Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland.
| |
Collapse
|
33
|
Mendes N, Dias Carvalho P, Martins F, Mendonça S, Malheiro AR, Ribeiro A, Carvalho J, Velho S. Animal Models to Study Cancer and Its Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:389-401. [PMID: 32130710 DOI: 10.1007/978-3-030-34025-4_20] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancers are complex tissues composed by genetically altered cancer cells and stromal elements such as inflammatory/immune cells, fibroblasts, endothelial cells and pericytes, neuronal cells, and a non-cellular component, the extracellular matrix. The complex network of interactions and crosstalk established between cancer cells and the supportig cellular and non-cellular components of the microenvironment are of extreme importance for tumor initiation and progression, strongly impacting the course and the outcome of the disease. Therefore, a better understanding of the tumorigenic processes implies the combined study of the cancer cell and the biologic, chemical and mechanic constituents of the tumor microenvironment, as their concerted action plays a major role in the carcinogenic pathway and is a key determinant of the efficacy of anti-cancer treatments. The use of animal models (e.g. Mouse, Zebrafish and Drosophila) to study cancer has greatly impacted our understanding of the processes governing initiation, progression and metastasis and allowed the discovery and pre-clinical validation of novel cancer treatments as it allows to recreate tumor development in a more pathophysiologic environment.
Collapse
Affiliation(s)
- N Mendes
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal.
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal.
| | - P Dias Carvalho
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - F Martins
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - S Mendonça
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - A R Malheiro
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IBMC, Instituto de Biologia Molecular e Celular da Universidade do Porto, Porto, Portugal
| | - A Ribeiro
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - J Carvalho
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal
| | - S Velho
- i3S, Instituto de Investigação e Inovação em Saúde, Porto, Portugal.
- IPATIMUP, Instituto de Patologia Molecular e Imunologia da Universidade do Porto, Porto, Portugal.
| |
Collapse
|
34
|
Yao Y, Wang L, Wang X. Modeling of Solid-Tumor Microenvironment in Zebrafish (Danio Rerio) Larvae. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:413-428. [PMID: 32130712 DOI: 10.1007/978-3-030-34025-4_22] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zebrafish larvae have emerged as a powerful model for studying tumorigenesis in vivo, with remarkable conservation with mammals in genetics, molecular and cell biology. Zebrafish tumor models bear the significant advantages of optical clarity in comparison to that in the mammalian models, allowing noninvasive investigation of the tumor cell and its microenvironment at single-cell resolution. Here we review recent progressions in the field of zebrafish models of solid tumor diseases in two main categories: the genetically engineered tumor models in which all cells in the tumor microenvironment are zebrafish cells, and xenograft tumor models in which the tumor microenvironment is composed of zebrafish cells and cells from other species. Notably, the zebrafish patient-derived xenograft (zPDX) models can be used for personalized drug assessment on primary tumor biopsies, including the pancreatic cancer. For the future studies, a series of high throughput drug screenings on the library of transgenic zebrafish models of solid tumor are expected to provide systematic database of oncogenic mutation, cell-of-origin, and leading compounds; and the humanization of zebrafish in genetics and cellular composition will make it more practical hosts for zPDX modeling. Together, zebrafish tumor model systems are unique and convenient in vivo platforms, with great potential to serve as valuable tools for cancer researches.
Collapse
Affiliation(s)
- Yuxiao Yao
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lei Wang
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xu Wang
- Cancer Metabolism Laboratory, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
ZeOncoTest: Refining and Automating the Zebrafish Xenograft Model for Drug Discovery in Cancer. Pharmaceuticals (Basel) 2019; 13:ph13010001. [PMID: 31878274 PMCID: PMC7169390 DOI: 10.3390/ph13010001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 12/12/2019] [Accepted: 12/23/2019] [Indexed: 12/24/2022] Open
Abstract
The xenograft of human cancer cells in model animals is a powerful tool for understanding tumor progression and metastatic potential. Mice represent a validated host, but their use is limited by the elevated experimental costs and low throughput. To overcome these restrictions, zebrafish larvae might represent a valuable alternative. Their small size and transparency allow the tracking of transplanted cells. Therefore, tumor growth and early steps of metastasis, which are difficult to evaluate in mice, can be addressed. In spite of its advantages, the use of this model has been hindered by lack of experimental homogeneity and validation. Considering these facts, the aim of our work was to standardize, automate, and validate a zebrafish larvae xenograft assay with increased translatability and higher drug screening throughput. The ZeOncoTest reliability is based on the optimization of different experimental parameters, such as cell labeling, injection site, automated individual sample image acquisition, and analysis. This workflow implementation finally allows a higher precision and experimental throughput increase, when compared to previous reports. The approach was validated with the breast cancer cell line MDA-MB-231, the colorectal cancer cells HCT116, and the prostate cancer cells PC3; and known drugs, respectively RKI-1447, Docetaxel, and Mitoxantrone. The results recapitulate growth and invasion for all tested tumor cells, along with expected efficacy of the compounds. Finally, the methodology has proven useful for understanding specific drugs mode of action. The insights gained bring a step further for zebrafish larvae xenografts to enter the regulated preclinical drug discovery path.
Collapse
|
36
|
Yang WY, Rao PS, Luo YC, Lin HK, Huang SH, Yang JM, Yuh CH. Omics-based Investigation of Diet-induced Obesity Synergized with HBx, Src, and p53 Mutation Accelerating Hepatocarcinogenesis in Zebrafish Model. Cancers (Basel) 2019; 11:cancers11121899. [PMID: 31795276 PMCID: PMC6966430 DOI: 10.3390/cancers11121899] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/17/2019] [Accepted: 11/25/2019] [Indexed: 12/19/2022] Open
Abstract
The primary type of liver cancer, hepatocellular carcinoma (HCC), has been associated with nonalcoholic steatohepatitis, diabetes, and obesity. Previous studies have identified some genetic risk factors, such as hepatitis B virus X antigens, overexpression of SRC oncogene, and mutation of the p53 tumor suppressor gene; however, the synergism between diet and genetic risk factors is still unclear. To investigate the synergism between diet and genetic risk factors in hepatocarcinogenesis, we used zebrafish with four genetic backgrounds and overfeeding or high-fat-diet-induced obesity with an omics-based expression of genes and histopathological changes. The results show that overfeeding and high-fat diet can induce obesity and nonalcoholic steatohepatitis in wild-type fish. In HBx, Src (p53-) triple transgenic zebrafish, diet-induced obesity accelerated HCC formation at five months of age and increased the cancer incidence threefold. We developed a global omics data analysis method to investigate genes, pathways, and biological systems based on microarray and next-generation sequencing (NGS, RNA-seq) omics data of zebrafish with four diet and genetic risk factors. The results show that two Kyoto Encyclopedia of Genes and Genomes (KEGG) systems, metabolism and genetic information processing, as well as the pathways of fatty acid metabolism, steroid biosynthesis, and ribosome biogenesis, are activated during hepatocarcinogenesis. This study provides a systematic view of the synergism between genetic and diet factors in the dynamic liver cancer formation process, and indicate that overfeeding or a high-fat diet and the risk genes have a synergistic effect in causing liver cancer by affecting fatty acid metabolism and ribosome biogenesis.
Collapse
Affiliation(s)
- Wan-Yu Yang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Miaoli, Taiwan; (W.-Y.Y.); (P.-S.R.); (H.-K.L.)
| | - Pei-Shu Rao
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Miaoli, Taiwan; (W.-Y.Y.); (P.-S.R.); (H.-K.L.)
- Department of Life Science, National Tsing-Hua University, Hsinchu 30070, Taiwan
| | - Yong-Chun Luo
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 30010, Taiwan; (Y.-C.L.); (S.-H.H.)
| | - Hua-Kuo Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Miaoli, Taiwan; (W.-Y.Y.); (P.-S.R.); (H.-K.L.)
| | - Sing-Han Huang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 30010, Taiwan; (Y.-C.L.); (S.-H.H.)
| | - Jinn-Moon Yang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu 30010, Taiwan; (Y.-C.L.); (S.-H.H.)
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-devices, National Chiao Tung University, Hsinchu 30010, Taiwan
- Correspondence: (J.-M.Y.); (C.-H.Y.); Tel.: +011-886-03-5712121*56942 (J.-M.Y.); +011-886-37-206166*35338 (C.-H.Y.)
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 35053, Miaoli, Taiwan; (W.-Y.Y.); (P.-S.R.); (H.-K.L.)
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan
- Institute of Bioinformatics and Structural Biology, National Tsing-Hua University, Hsinchu 30070, Taiwan
- Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (J.-M.Y.); (C.-H.Y.); Tel.: +011-886-03-5712121*56942 (J.-M.Y.); +011-886-37-206166*35338 (C.-H.Y.)
| |
Collapse
|
37
|
Hason M, Bartůněk P. Zebrafish Models of Cancer-New Insights on Modeling Human Cancer in a Non-Mammalian Vertebrate. Genes (Basel) 2019; 10:genes10110935. [PMID: 31731811 PMCID: PMC6896156 DOI: 10.3390/genes10110935] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/11/2019] [Accepted: 11/11/2019] [Indexed: 12/26/2022] Open
Abstract
Zebrafish (Danio rerio) is a valuable non-mammalian vertebrate model widely used to study development and disease, including more recently cancer. The evolutionary conservation of cancer-related programs between human and zebrafish is striking and allows extrapolation of research outcomes obtained in fish back to humans. Zebrafish has gained attention as a robust model for cancer research mainly because of its high fecundity, cost-effective maintenance, dynamic visualization of tumor growth in vivo, and the possibility of chemical screening in large numbers of animals at reasonable costs. Novel approaches in modeling tumor growth, such as using transgene electroporation in adult zebrafish, could improve our knowledge about the spatial and temporal control of cancer formation and progression in vivo. Looking at genetic as well as epigenetic alterations could be important to explain the pathogenesis of a disease as complex as cancer. In this review, we highlight classic genetic and transplantation models of cancer in zebrafish as well as provide new insights on advances in cancer modeling. Recent progress in zebrafish xenotransplantation studies and drug screening has shown that zebrafish is a reliable model to study human cancer and could be suitable for evaluating patient-derived xenograft cell invasiveness. Rapid, large-scale evaluation of in vivo drug responses and kinetics in zebrafish could undoubtedly lead to new applications in personalized medicine and combination therapy. For all of the above-mentioned reasons, zebrafish is approaching a future of being a pre-clinical cancer model, alongside the mouse. However, the mouse will continue to be valuable in the last steps of pre-clinical drug screening, mostly because of the highly conserved mammalian genome and biological processes.
Collapse
|
38
|
Tomko A, O'Leary L, Trask H, Achenbach JC, Hall SR, Goralski KB, Ellis LD, Dupré DJ. Antitumor Activity of Abnormal Cannabidiol and Its Analog O-1602 in Taxol-Resistant Preclinical Models of Breast Cancer. Front Pharmacol 2019; 10:1124. [PMID: 31611800 PMCID: PMC6777324 DOI: 10.3389/fphar.2019.01124] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/30/2019] [Indexed: 12/31/2022] Open
Abstract
Cannabinoids exhibit anti-inflammatory and antitumorigenic properties. Contrary to most cannabinoids present in the Cannabis plant, some, such as O-1602 and abnormal cannabidiol, have no or only little affinity to the CB1 or CB2 cannabinoid receptors and instead exert their effects through other receptors. Here, we investigated whether the synthetic regioisomers of cannabidiol, abnormal cannabidiol, and a closely related compound, O-1602, display antitumorigenic effects in cellular models of breast cancer and whether it could reduce tumorigenesis in vivo. Several studies have shown the effects of cannabinoids on chemotherapy-sensitive breast cancer cell lines, but less is known about the antitumorigenic effects of cannabinoids in chemotherapy-resistant cell lines. Paclitaxel-resistant MDA-MB-231 and MCF-7 breast cancer cell lines were used to study the effect of O-1602 and abnormal cannabidiol on viability, apoptosis, and migration. The effects of O-1602 and abnormal cannabidiol on cell viability were completely blocked by the combination of GPR55 and GPR18-specific siRNAs. Both O-1602 and abnormal cannabidiol decreased viability in paclitaxel-resistant breast cancer cells in a concentration-dependent manner through induction of apoptosis. The effect of these cannabinoids on tumor growth in vivo was studied in a zebrafish xenograft model. In this model, treatment with O-1602 and abnormal cannabidiol (2 µM) significantly reduced tumor growth. Our results suggest that atypical cannabinoids, like O-1602 and abnormal cannabidiol, exert antitumorigenic effects on paclitaxel-resistant breast cancer cells. Due to their lack of central sedation and psychoactive effects, these atypical cannabinoids could represent new leads for the development of additional anticancer treatments when resistance to conventional chemotherapy occurs during the treatment of breast and possibly other cancers.
Collapse
Affiliation(s)
- Andrea Tomko
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Lauren O'Leary
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - Hilary Trask
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| | - John C Achenbach
- Aquatic and Crop Resource Development Research Center, National Research Council of Canada, Halifax, Canada
| | - Steven R Hall
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada.,College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, Canada
| | - Kerry B Goralski
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada.,College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, Canada
| | - Lee D Ellis
- College of Pharmacy, Faculty of Health, Dalhousie University, Halifax, Canada
| | - Denis J Dupré
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, Canada
| |
Collapse
|
39
|
Su ZL, Su CW, Huang YL, Yang WY, Sampurna BP, Ouchi T, Lee KL, Wu CS, Wang HD, Yuh CH. A Novel AURKA Mutant-Induced Early-Onset Severe Hepatocarcinogenesis Greater than Wild-Type via Activating Different Pathways in Zebrafish. Cancers (Basel) 2019; 11:cancers11070927. [PMID: 31269749 PMCID: PMC6678475 DOI: 10.3390/cancers11070927] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 12/18/2022] Open
Abstract
Aurora A kinase (AURKA) is an important regulator in mitotic progression and is overexpressed frequently in human cancers, including hepatocellular carcinoma (HCC). Many AURKA mutations were identified in cancer patients. Overexpressing wild-type Aurka developed a low incidence of hepatic tumors after long latency in mice. However, none of the AURKA mutant animal models have ever been described. The mechanism of mutant AURKA-mediated hepatocarcinogenesis is still unclear. A novel AURKA mutation with a.a.352 Valine to Isoleucine (V352I) was identified from clinical specimens. By using liver-specific transgenic fish overexpressing both the mutant and wild-type AURKA, the AURKA(V352I)-induced hepatocarcinogenesis was earlier and much more severe than wild-type AURKA. Although an increase of the expression of lipogenic enzyme and lipogenic factor was observed in both AURKA(V352I) and AURKA(WT) transgenic fish, AURKA(V352I) has a greater probability to promote fibrosis at 3 months compared to AURKA(WT). Furthermore, the expression levels of cell cycle/proliferation markers were higher in the AURKA(V352I) mutant than AURKA(WT) in transgenic fish, implying that the AURKA(V352I) mutant may accelerate HCC progression. Moreover, we found that the AURKA(V352I) mutant activates AKT signaling and increases nuclear β-catenin, but AURKA(WT) only activates membrane form β-catenin, which may account for the differences. In this study, we provide a new insight, that the AURKA(V352I) mutation contributes to early onset hepatocarcinogenesis, possibly through activation of different pathways than AURKA(WT). This transgenic fish may serve as a drug-screening platform for potential precision medicine therapeutics.
Collapse
Affiliation(s)
- Zhong-Liang Su
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chien-Wei Su
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yi-Luen Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Wan-Yu Yang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Bonifasius Putera Sampurna
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan
| | - Toru Ouchi
- Department of Cancer Genetics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Kuan-Lin Lee
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chen-Sheng Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Horng-Dar Wang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan.
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu 30010, Taiwan.
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
40
|
Proietti S, Cucina A, Pensotti A, Biava PM, Minini M, Monti N, Catizone A, Ricci G, Leonetti E, Harrath AH, Alwasel SH, Bizzarri M. Active Fraction from Embryo Fish Extracts Induces Reversion of the Malignant Invasive Phenotype in Breast Cancer through Down-regulation of TCTP and Modulation of E-cadherin/β-catenin Pathway. Int J Mol Sci 2019; 20:E2151. [PMID: 31052313 PMCID: PMC6539734 DOI: 10.3390/ijms20092151] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/15/2019] [Accepted: 04/29/2019] [Indexed: 12/01/2022] Open
Abstract
Some yet unidentified factors released by both oocyte and embryonic microenvironments demonstrated to be non-permissive for tumor development and display the remarkable ability to foster cell/tissue reprogramming, thus ultimately reversing the malignant phenotype. In the present study we observed how molecular factors extracted from Zebrafish embryos during specific developmental phases (20 somites) significantly antagonize proliferation of breast cancer cells, while reversing a number of prominent aspects of malignancy. Embryo extracts reduce cell proliferation, enhance apoptosis, and dramatically inhibit both invasiveness and migrating capabilities of cancer cells. Counteracting the invasive phenotype is a relevant issue in controlling tumor spreading and metastasis. Moreover, such effect is not limited to cancerous cells as embryo extracts were also effective in inhibiting migration and invasiveness displayed by normal breast cells undergoing epithelial-mesenchymal transition upon TGF-β1 stimulation. The reversion program involves the modulation of E-cadherin/β-catenin pathway, cytoskeleton remodeling with dramatic reduction in vinculin, as well as downregulation of TCTP and the concomitant increase in p53 levels. Our findings highlight that-contrary to the prevailing current "dogma", which posits that neoplastic cells are irreversibly "committed"-the malignant phenotype can ultimately be "reversed", at least partially, in response to environmental morphogenetic influences.
Collapse
Affiliation(s)
- Sara Proietti
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, 00161 Rome, Italy.
| | - Alessandra Cucina
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, 00161 Rome, Italy.
- Azienda Policlinico Umberto I, 00161 Rome, Italy.
| | | | - Pier Mario Biava
- Scientific Institute of Research and Health Care (IRCCS) Multimedica, 20099 Milano, Italy.
| | - Mirko Minini
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, 00161 Rome, Italy.
- Department of Experimental Medicine, Sapienza University of Rome, Systems Biology Group Lab, 00161 Rome, Italy.
| | - Noemi Monti
- Department of Surgery "Pietro Valdoni", Sapienza University of Rome, 00161 Rome, Italy.
| | - Angela Catizone
- Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy.
| | - Giulia Ricci
- Department. of Experimental Medicine, Università degli Studi della Campania "Luigi Vanvitelli", 80138 Naples, Italy.
| | - Erica Leonetti
- Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy.
| | - Abdel Halim Harrath
- Department of Zoology, College of Science, King Saud University, Riyadh 2455, Saudi Arabia.
| | - Saleh H Alwasel
- Department of Zoology, College of Science, King Saud University, Riyadh 2455, Saudi Arabia.
| | - Mariano Bizzarri
- Department of Anatomy, Histology, Forensic-Medicine and Orthopedics, "Sapienza" University of Rome, 00161 Rome, Italy.
| |
Collapse
|
41
|
Lu JW, Raghuram D, Fong PSA, Gong Z. Inducible Intestine-Specific Expression of kras V12 Triggers Intestinal Tumorigenesis In Transgenic Zebrafish. Neoplasia 2018; 20:1187-1197. [PMID: 30390498 PMCID: PMC6215966 DOI: 10.1016/j.neo.2018.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
KRAS mutations are a major risk factor in colorectal cancers. In particular, a point mutation of KRAS of amino acid 12, such as KRASV12, renders it stable activity in oncogenesis. We found that krasV12 promotes intestinal carcinogenesis by generating a transgenic zebrafish line with inducible krasV12 expression in the intestine, Tg(ifabp:EGFP-krasV12). The transgenic fish generated exhibited significant increases in the rates of intestinal epithelial outgrowth, proliferation, and cross talk in the active Ras signaling pathway involving in epithelial-mesenchymal transition (EMT). These results provide in vivo evidence of Ras pathway activation via krasV12 overexpression. Long-term transgenic expression of krasV12 resulted in enteritis, epithelial hyperplasia, and tubular adenoma in adult fish. This was accompanied by increased levels of the signaling proteins p-Erk and p-Akt and by downregulation of the EMT marker E-cadherin. Furthermore, we also observed a synergistic effect of krasV12 expression and dextran sodium sulfate treatment to enhance intestinal tumor in zebrafish. Our results demonstrate that krasV12 overexpression induces intestinal tumorigenesis in zebrafish, which mimics intestinal tumor formation in humans. Thus, our transgenic zebrafish may provide a valuable in vivo platform that can be used to investigate tumor initiation and anticancer drugs for gastrointestinal cancers.
Collapse
Affiliation(s)
- Jeng-Wei Lu
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Divya Raghuram
- Department of Biological Sciences, National University of Singapore, Singapore
| | | | - Zhiyuan Gong
- Department of Biological Sciences, National University of Singapore, Singapore.
| |
Collapse
|
42
|
Dobson THW, Gopalakrishnan V. Preclinical Models of Pediatric Brain Tumors-Forging Ahead. Bioengineering (Basel) 2018; 5:bioengineering5040081. [PMID: 30279402 PMCID: PMC6315787 DOI: 10.3390/bioengineering5040081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/22/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022] Open
Abstract
Approximately five out of 100,000 children from 0 to 19 years old are diagnosed with a brain tumor. These children are treated with medication designed for adults that are highly toxic to a developing brain. Those that survive are at high risk for a lifetime of limited physical, psychological, and cognitive abilities. Despite much effort, not one drug exists that was designed specifically for pediatric patients. Stagnant government funding and the lack of economic incentives for the pharmaceutical industry greatly limits preclinical research and the development of clinically applicable pediatric brain tumor models. As more data are collected, the recognition of disease sub-groups based on molecular heterogeneity increases the need for designing specific models suitable for predictive drug screening. To overcome these challenges, preclinical approaches will need continual enhancement. In this review, we examine the advantages and shortcomings of in vitro and in vivo preclinical pediatric brain tumor models and explore potential solutions based on past, present, and future strategies for improving their clinical relevancy.
Collapse
Affiliation(s)
- Tara H W Dobson
- Department of Pediatrics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | - Vidya Gopalakrishnan
- Department of Pediatrics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Department of Molecular & Cellular Oncology, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Brain Tumor Center, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Center for Cancer Epigenetics, University of Texas, M.D. Anderson Cancer Center, Houston, TX 77030, USA.
- Graduate School of Biomedical Sciences UT-Health Science Center, Houston, TX 77030, USA.
| |
Collapse
|
43
|
Sturtzel C, Lipnik K, Hofer-Warbinek R, Testori J, Ebner B, Seigner J, Qiu P, Bilban M, Jandrositz A, Preisegger KH, Untergasser G, Gunsilius E, de Martin R, Kroll J, Hofer E. FOXF1 Mediates Endothelial Progenitor Functions and Regulates Vascular Sprouting. Front Bioeng Biotechnol 2018; 6:76. [PMID: 29963552 PMCID: PMC6010557 DOI: 10.3389/fbioe.2018.00076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/24/2018] [Indexed: 01/26/2023] Open
Abstract
Endothelial colony forming cells (ECFC) or late blood outgrowth endothelial cells (BOEC) have been proposed to contribute to neovascularization in humans. Exploring genes characteristic for the progenitor status of ECFC we have identified the forkhead box transcription factor FOXF1 to be selectively expressed in ECFC compared to mature endothelial cells isolated from the vessel wall. Analyzing the role of FOXF1 by gain- and loss-of-function studies we detected a strong impact of FOXF1 expression on the particularly high sprouting capabilities of endothelial progenitors. This apparently relates to the regulation of expression of several surface receptors. First, FOXF1 overexpression specifically induces the expression of Notch2 receptors and induces sprouting. Vice versa, knock-down of FOXF1 and Notch2 reduces sprouting. In addition, FOXF1 augments the expression of VEGF receptor-2 and of the arterial marker ephrin B2, whereas it downmodulates the venous marker EphB4. In line with these findings on human endothelial progenitors, we further show that knockdown of FOXF1 in the zebrafish model alters, during embryonic development, the regular formation of vasculature by sprouting. Hence, these findings support a crucial role of FOXF1 for endothelial progenitors and connected vascular sprouting as it may be relevant for tissue neovascularization. It further implicates Notch2, VEGF receptor-2, and ephrin B2 as downstream mediators of FOXF1 functions.
Collapse
Affiliation(s)
- Caterina Sturtzel
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Karoline Lipnik
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Renate Hofer-Warbinek
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Julia Testori
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Bettina Ebner
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jaqueline Seigner
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ping Qiu
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine & Core Facility Genomics, Core Facilities, Medical University of Vienna, Vienna, Austria
| | | | - Karl-Heinz Preisegger
- VivoCell Biosolutions GmbH, Graz, Austria.,Institut für morphologische Analytik und Humangenetik, Graz, Austria
| | - Gerold Untergasser
- Laboratory for Tumor Biology & Angiogenesis, Medical University of Innsbruck, Innsbruck, Austria
| | - Eberhard Gunsilius
- Laboratory for Tumor Biology & Angiogenesis, Medical University of Innsbruck, Innsbruck, Austria
| | - Rainer de Martin
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Jens Kroll
- Department of Vascular Biology and Tumor Angiogenesis, European for Center for Angioscience, Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Erhard Hofer
- Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
44
|
Affiliation(s)
- Jason R. Meyers
- Department of Biology and Program in Neuroscience, Colgate University; Hamilton New York
| |
Collapse
|
45
|
Gutiérrez-Lovera C, Vázquez-Ríos AJ, Guerra-Varela J, Sánchez L, de la Fuente M. The Potential of Zebrafish as a Model Organism for Improving the Translation of Genetic Anticancer Nanomedicines. Genes (Basel) 2017; 8:E349. [PMID: 29182542 PMCID: PMC5748667 DOI: 10.3390/genes8120349] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/06/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022] Open
Abstract
In the last few decades, the field of nanomedicine applied to cancer has revolutionized cancer treatment: several nanoformulations have already reached the market and are routinely being used in the clinical practice. In the case of genetic nanomedicines, i.e., designed to deliver gene therapies to cancer cells for therapeutic purposes, advances have been less impressive. This is because of the many barriers that limit the access of the therapeutic nucleic acids to their target site, and the lack of models that would allow for an improvement in the understanding of how nanocarriers can be tailored to overcome them. Zebrafish has important advantages as a model species for the study of anticancer therapies, and have a lot to offer regarding the rational development of efficient delivery of genetic nanomedicines, and hence increasing the chances of their successful translation. This review aims to provide an overview of the recent advances in the development of genetic anticancer nanomedicines, and of the zebrafish models that stand as promising tools to shed light on their mechanisms of action and overall potential in oncology.
Collapse
Affiliation(s)
- C Gutiérrez-Lovera
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| | - A J Vázquez-Ríos
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| | - J Guerra-Varela
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
- Geneaqua S.L., Lugo 27002, Spain.
| | - L Sánchez
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
| | - M de la Fuente
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| |
Collapse
|