1
|
Zhou Z, Sun Y, Yang J, Abliz Z. Mapping the Metabolic Characteristics and Perturbation of Adult Casper Zebrafish by Ambient Mass Spectrometry Imaging. Metabolites 2024; 14:204. [PMID: 38668332 PMCID: PMC11051737 DOI: 10.3390/metabo14040204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Casper, a type of transparent mutant-line zebrafish, was generated to overcome the opaque trunk of an adult zebrafish for tumor modeling to realize real-time visualization of transplanted cells in vivo. However, the molecular information at the metabolic level has not received much attention. Herein, a spatially resolved metabolomics method based on an airflow-assisted desorption electrospray ionization-mass spectrometry imaging (AFADESI-MSI) system for whole-body zebrafish was used to investigate small molecules and the distribution of adult casper (Mitfaw2/w2, roya9/a9) and the differences from wild-type zebrafish. Finally, the spatial distribution information of more than 1500 endogenous ions was obtained in positive and negative detection modes, and 186 metabolites belonging to a variety of structural categories were identified or annotated. Compared with wild-type samples, 85 variables, including 37 known metabolites, were screened out. In addition, the disordered metabolic pathways caused by the genetic mutation were excavated, involving downregulation of purine metabolism and arachidonic acid metabolism, upregulation of glycerophospholipid metabolism, and biosynthesis of unsaturated fatty acids. All these results were observed in the most intuitive way through MSI. This study revealed important metabolic characteristics of and perturbation in adult casper zebrafish, and provides indispensable fundamental knowledge for tumor research based on it.
Collapse
Affiliation(s)
- Zhi Zhou
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China;
- College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China; (Y.S.); (J.Y.)
| | - Yue Sun
- College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China; (Y.S.); (J.Y.)
| | - Ji Yang
- College of Life and Environmental Sciences, Minzu University of China, 27 Zhongguancun South Avenue, Beijing 100081, China; (Y.S.); (J.Y.)
| | - Zeper Abliz
- Key Laboratory of Mass Spectrometry Imaging and Metabolomics (Minzu University of China), National Ethnic Affairs Commission, Beijing 100081, China;
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
2
|
Roy D, Subramaniam B, Chong WC, Bornhorst M, Packer RJ, Nazarian J. Zebrafish-A Suitable Model for Rapid Translation of Effective Therapies for Pediatric Cancers. Cancers (Basel) 2024; 16:1361. [PMID: 38611039 PMCID: PMC11010887 DOI: 10.3390/cancers16071361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Pediatric cancers are the leading cause of disease-related deaths in children and adolescents. Most of these tumors are difficult to treat and have poor overall survival. Concerns have also been raised about drug toxicity and long-term detrimental side effects of therapies. In this review, we discuss the advantages and unique attributes of zebrafish as pediatric cancer models and their importance in targeted drug discovery and toxicity assays. We have also placed a special focus on zebrafish models of pediatric brain cancers-the most common and difficult solid tumor to treat.
Collapse
Affiliation(s)
- Debasish Roy
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Bavani Subramaniam
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Wai Chin Chong
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Miriam Bornhorst
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Roger J. Packer
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
| | - Javad Nazarian
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC 20012, USA; (D.R.)
- DIPG/DMG Research Center Zurich, Children’s Research Center, Department of Pediatrics, University Children’s Hospital Zürich, 8032 Zurich, Switzerland
| |
Collapse
|
3
|
Senrung A, Tripathi T, Aggarwal N, Janjua D, Yadav J, Chaudhary A, Chhokar A, Joshi U, Bharti AC. Phytochemicals Showing Antiangiogenic Effect in Pre-clinical Models and their Potential as an Alternative to Existing Therapeutics. Curr Top Med Chem 2024; 24:259-300. [PMID: 37867279 DOI: 10.2174/0115680266264349231016094456] [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: 05/27/2023] [Revised: 07/25/2023] [Accepted: 08/10/2023] [Indexed: 10/24/2023]
Abstract
Angiogenesis, the formation of new blood vessels from a pre-existing vascular network, is an important hallmark of several pathological conditions, such as tumor growth and metastasis, proliferative retinopathies, including proliferative diabetic retinopathy and retinopathy of prematurity, age-related macular degeneration, rheumatoid arthritis, psoriasis, and endometriosis. Putting a halt to pathology-driven angiogenesis is considered an important therapeutic strategy to slow down or reduce the severity of pathological disorders. Considering the attrition rate of synthetic antiangiogenic compounds from the lab to reaching the market due to severe side effects, several compounds of natural origin are being explored for their antiangiogenic properties. Employing pre-clinical models for the evaluation of novel antiangiogenic compounds is a promising strategy for rapid screening of antiangiogenic compounds. These studies use a spectrum of angiogenic model systems that include HUVEC two-dimensional culture, nude mice, chick chorioallantoic membrane, transgenic zebrafish, and dorsal aorta from rats and chicks, depending upon available resources. The present article emphasizes the antiangiogenic activity of the phytochemicals shown to exhibit antiangiogenic behavior in these well-defined existing angiogenic models and highlights key molecular targets. Different models help to get a quick understanding of the efficacy and therapeutics mechanism of emerging lead molecules. The inherent variability in assays and corresponding different phytochemicals tested in each study prevent their immediate utilization in clinical studies. This review will discuss phytochemicals discovered using suitable preclinical antiangiogenic models, along with a special mention of leads that have entered clinical evaluation.
Collapse
Affiliation(s)
- Anna Senrung
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Neuropharmacology and Drug Delivery Laboratory, Daulat Ram College, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Nikita Aggarwal
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Divya Janjua
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Joni Yadav
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Apoorva Chaudhary
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Arun Chhokar
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Deshbandhu College, University of Delhi, Delhi, India
| | - Udit Joshi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Alok Chandra Bharti
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| |
Collapse
|
4
|
Al-Hamaly MA, Turner LT, Rivera-Martinez A, Rodriguez A, Blackburn JS. Zebrafish Cancer Avatars: A Translational Platform for Analyzing Tumor Heterogeneity and Predicting Patient Outcomes. Int J Mol Sci 2023; 24:2288. [PMID: 36768609 PMCID: PMC9916713 DOI: 10.3390/ijms24032288] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
The increasing number of available anti-cancer drugs presents a challenge for oncologists, who must choose the most effective treatment for the patient. Precision cancer medicine relies on matching a drug with a tumor's molecular profile to optimize the therapeutic benefit. However, current precision medicine approaches do not fully account for intra-tumoral heterogeneity. Different mutation profiles and cell behaviors within a single heterogeneous tumor can significantly impact therapy response and patient outcomes. Patient-derived avatar models recapitulate a patient's tumor in an animal or dish and provide the means to functionally assess heterogeneity's impact on drug response. Mouse xenograft and organoid avatars are well-established, but the time required to generate these models is not practical for clinical decision-making. Zebrafish are emerging as a time-efficient and cost-effective cancer avatar model. In this review, we highlight recent developments in zebrafish cancer avatar models and discuss the unique features of zebrafish that make them ideal for the interrogation of cancer heterogeneity and as part of precision cancer medicine pipelines.
Collapse
Affiliation(s)
- Majd A. Al-Hamaly
- Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40356, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
| | - Logan T. Turner
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, USA
| | | | - Analiz Rodriguez
- Department of Neurosurgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jessica S. Blackburn
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA
- Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40356, USA
| |
Collapse
|
5
|
Engraftment of Allotransplanted Tumor Cells in Adult rag2 Mutant Xenopus tropicalis. Cancers (Basel) 2022; 14:cancers14194560. [PMID: 36230482 PMCID: PMC9559464 DOI: 10.3390/cancers14194560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary While the mouse is without doubt the most studied animal for experimental cancer research, aquatic vertebrates such as zebrafish have also contributed to the field. More recently, thanks to the Nobel-prize winning technology of CRISPR/Cas mediated genomic engineering, the frog Xenopus tropicalis has emerged as an additional powerful model for studying human cancer. Via CRISPR-mediated genome editing, several models for different human cancers have been obtained in this animal. However, what has been lacking in Xenopus is the possibility to transplant tumor cells between different frogs. This is important to allow better characterization of the tumor cells and exploration of therapeutic opportunities. In this paper, we describe the generation of a genetic mutant in Xenopus tropicalis that has a compromised immune system, thereby allowing the grafting and expansion of tumors obtained in this species. In addition, an optimized protocol is provided for the irradiation of wild-type Xenopus frogs that subsequently are temporarily immunocompromised and during that period allow tumor engraftment. This work will expand the toolbox for modeling human cancer in Xenopus tropicalis, thereby further establishing it as a powerful experimental cancer model. Abstract Modeling human genetic diseases and cancer in lab animals has been greatly aided by the emergence of genetic engineering tools such as TALENs and CRISPR/Cas9. We have previously demonstrated the ease with which genetically engineered Xenopus models (GEXM) can be generated via injection of early embryos with Cas9 recombinant protein loaded with sgRNAs targeting single or multiple tumor suppressor genes. What has been lacking so far is the possibility to propagate and characterize the induced cancers via transplantation. Here, we describe the generation of a rag2 knockout line in Xenopus tropicalis that is deficient in functional T and B cells. This line was validated by means of allografting experiments with primary tp53−/− and apc+/−/tp53−/− donor tumors. In addition, we optimized available protocols for the sub-lethal irradiation of wild-type X. tropicalis froglets. Irradiated animals also allowed the stable, albeit transient, engraftment of transplanted X. tropicalis tumor cells. The novel rag2−/− line and the irradiated wild-type froglets will further expand the experimental toolbox in the diploid amphibian X. tropicalis and help to establish it as a versatile and relevant model for exploring human cancer.
Collapse
|
6
|
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
|
7
|
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
|
8
|
Yan C, Yang Q, Zhang S, Millar DG, Alpert EJ, Do D, Veloso A, Brunson DC, Drapkin BJ, Stanzione M, Scarfò I, Moore JC, Iyer S, Qin Q, Wei Y, McCarthy KM, Rawls JF, Dyson NJ, Cobbold M, Maus MV, Langenau DM. Single-cell imaging of T cell immunotherapy responses in vivo. J Exp Med 2021; 218:e20210314. [PMID: 34415995 PMCID: PMC8383813 DOI: 10.1084/jem.20210314] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/19/2021] [Accepted: 07/09/2021] [Indexed: 12/22/2022] Open
Abstract
T cell immunotherapies have revolutionized treatment for a subset of cancers. Yet, a major hurdle has been the lack of facile and predicative preclinical animal models that permit dynamic visualization of T cell immune responses at single-cell resolution in vivo. Here, optically clear immunocompromised zebrafish were engrafted with fluorescent-labeled human cancers along with chimeric antigen receptor T (CAR T) cells, bispecific T cell engagers (BiTEs), and antibody peptide epitope conjugates (APECs), allowing real-time single-cell visualization of T cell-based immunotherapies in vivo. This work uncovered important differences in the kinetics of T cell infiltration, tumor cell engagement, and killing between these immunotherapies and established early endpoint analysis to predict therapy responses. We also established EGFR-targeted immunotherapies as a powerful approach to kill rhabdomyosarcoma muscle cancers, providing strong preclinical rationale for assessing a wider array of T cell immunotherapies in this disease.
Collapse
Affiliation(s)
- Chuan Yan
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Qiqi Yang
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Songfa Zhang
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - David G. Millar
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - Eric J. Alpert
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Daniel Do
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Alexandra Veloso
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Dalton C. Brunson
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Benjamin J. Drapkin
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - Marcello Stanzione
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - Irene Scarfò
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - John C. Moore
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Sowmya Iyer
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Qian Qin
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Yun Wei
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - Karin M. McCarthy
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC
| | - Nick J. Dyson
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - Mark Cobbold
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Early Oncology R&D, AstraZeneca, Gaithersburg, MD
| | - Marcela V. Maus
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
| | - David M. Langenau
- Molecular Pathology Unit, Massachusetts General Research Institute, Charlestown, MA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Harvard Stem Cell Institute, Cambridge, MA
| |
Collapse
|
9
|
Pensado-López A, Fernández-Rey J, Reimunde P, Crecente-Campo J, Sánchez L, Torres Andón F. Zebrafish Models for the Safety and Therapeutic Testing of Nanoparticles with a Focus on Macrophages. NANOMATERIALS 2021; 11:nano11071784. [PMID: 34361170 PMCID: PMC8308170 DOI: 10.3390/nano11071784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022]
Abstract
New nanoparticles and biomaterials are increasingly being used in biomedical research for drug delivery, diagnostic applications, or vaccines, and they are also present in numerous commercial products, in the environment and workplaces. Thus, the evaluation of the safety and possible therapeutic application of these nanomaterials has become of foremost importance for the proper progress of nanotechnology. Due to economical and ethical issues, in vitro and in vivo methods are encouraged for the testing of new compounds and/or nanoparticles, however in vivo models are still needed. In this scenario, zebrafish (Danio rerio) has demonstrated potential for toxicological and pharmacological screenings. Zebrafish presents an innate immune system, from early developmental stages, with conserved macrophage phenotypes and functions with respect to humans. This fact, combined with the transparency of zebrafish, the availability of models with fluorescently labelled macrophages, as well as a broad variety of disease models offers great possibilities for the testing of new nanoparticles. Thus, with a particular focus on macrophage-nanoparticle interaction in vivo, here, we review the studies using zebrafish for toxicological and biodistribution testing of nanoparticles, and also the possibilities for their preclinical evaluation in various diseases, including cancer and autoimmune, neuroinflammatory, and infectious diseases.
Collapse
Affiliation(s)
- Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Juan Fernández-Rey
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Pedro Reimunde
- Department of Physiotherapy, Medicine and Biomedical Sciences, Universidade da Coruña, Campus de Oza, 15006 A Coruña, Spain;
- Department of Neurosurgery, Hospital Universitario Lucus Augusti, 27003 Lugo, Spain
| | - José Crecente-Campo
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (J.F.-R.)
- Correspondence: (L.S.); (F.T.A.)
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
- Correspondence: (L.S.); (F.T.A.)
| |
Collapse
|
10
|
Lin FJ, Li H, Wu DT, Zhuang QG, Li HB, Geng F, Gan RY. Recent development in zebrafish model for bioactivity and safety evaluation of natural products. Crit Rev Food Sci Nutr 2021; 62:8646-8674. [PMID: 34058920 DOI: 10.1080/10408398.2021.1931023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zebrafish is a species of freshwater fish, popular in aquariums and laboratories. Several advantageous features have facilitated zebrafish to be extensively utilized as a valuable vertebrate model in the lab. It has been well-recognized that natural products possess multiple health benefits for humans. With the increasing demand for natural products in the development of functional foods, nutraceuticals, and natural cosmetics, the zebrafish has emerged as an unprecedented tool for rapidly and economically screening and identifying safe and effective substances from natural products. This review first summarized the key factors for the management of zebrafish in the laboratory, followed by highlighting the current progress on the establishment and applications of zebrafish models in the bioactivity evaluation of natural products. In addition, the zebrafish models used for assessing the potential toxicity or health risks of natural products were involved as well. Overall, this review indicates that zebrafish are promising animal models for the bioactivity and safety evaluation of natural products, and zebrafish models can accelerate the discovery of novel natural products with potential health functions.
Collapse
Affiliation(s)
- Fang-Jun Lin
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Hang Li
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| | - Ding-Tao Wu
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Qi-Guo Zhuang
- China-New Zealand Belt and Road Joint Laboratory on Kiwifruit, Sichuan Provincial Academy of Natural Resource Sciences, Chengdu, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Ren-You Gan
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| |
Collapse
|
11
|
Lipsitt A, Hensch NR, Moreno-Campos R, Baxi K, Chen J, Challa AK, Chen EY, Ignatius MS. Zebrafish Tumor Graft Transplantation to Grow Tumors In Vivo That Engraft Poorly as Single Cell Suspensions. Zebrafish 2021; 18:293-296. [PMID: 34030492 DOI: 10.1089/zeb.2021.0006] [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] [Indexed: 11/12/2022] Open
Abstract
Angiosarcoma is a clinically aggressive tumor with a high rate of mortality. It can arise in vascular or lymphatic tissues, involve any part of the body, and aggressively spread locally or metastasize. Angiosarcomas spontaneously develop in the tp53 deleted (tp53del/del) zebrafish mutant. However, established protocols for tumor dissection and transplantation of single cell suspensions of angiosarcoma tumors result in inferior implantation rates. To resolve these complications, we developed a new tumor grafting technique for engraftment of angiosarcoma and similar tumors in zebrafish, which maintains the tumor microenvironment and has superior rates of engraftment.
Collapse
Affiliation(s)
- Amanda Lipsitt
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
- Department of Pediatrics, Division of Hematology Oncology, UT Health Sciences Center, San Antonio, Texas, USA
| | - Nicole R Hensch
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
| | - Rodrigo Moreno-Campos
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
| | - Kunal Baxi
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
| | - Jiangfei Chen
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
- Institute of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, P.R. China
| | - Anil K Challa
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Eleanor Y Chen
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Myron S Ignatius
- Department of Molecular Medicine, Greehey Children's Cancer Research Institute (GCCRI), UT Health Sciences Center, San Antonio, Texas, USA
| |
Collapse
|
12
|
Rong J, He Y, Tang J, Qiao R, Lin S. "Fishing" nano-bio interactions at the key biological barriers. NANOSCALE 2021; 13:5954-5964. [PMID: 33734277 DOI: 10.1039/d1nr00328c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Understanding nano-bio interactions is pivotal to the safe implementation of nanotechnology for both biological and environmental applications. Zebrafish as a model organism provides unique opportunities to dissect nano-bio interactions occurring at different biological barriers. In this review, we focus on four key biological barriers, namely cell membrane, blood-brain barrier (BBB), skin and gill epithelia, and gastrointestinal tract (GIT), and highlight recent advancement achieved by using zebrafish to conduct both visualized observations and mechanistic investigations on a diversity of nano-bio interactions.
Collapse
Affiliation(s)
- Jinyu Rong
- College of Environmental Science and Engineering, Biomedical Multidisciplinary Innovation Research Institute, Shanghai East Hospital, Tongji University, Shanghai 200092, China.
| | | | | | | | | |
Collapse
|
13
|
Reimunde P, Pensado-López A, Carreira Crende M, Lombao Iglesias V, Sánchez L, Torrecilla-Parra M, Ramírez CM, Anfray C, Torres Andón F. Cellular and Molecular Mechanisms Underlying Glioblastoma and Zebrafish Models for the Discovery of New Treatments. Cancers (Basel) 2021; 13:1087. [PMID: 33802571 PMCID: PMC7961726 DOI: 10.3390/cancers13051087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common of all brain malignant tumors; it displays a median survival of 14.6 months with current complete standard treatment. High heterogeneity, aggressive and invasive behavior, the impossibility of completing tumor resection, limitations for drug administration and therapeutic resistance to current treatments are the main problems presented by this pathology. In recent years, our knowledge of GBM physiopathology has advanced significantly, generating relevant information on the cellular heterogeneity of GBM tumors, including cancer and immune cells such as macrophages/microglia, genetic, epigenetic and metabolic alterations, comprising changes in miRNA expression. In this scenario, the zebrafish has arisen as a promising animal model to progress further due to its unique characteristics, such as transparency, ease of genetic manipulation, ethical and economic advantages and also conservation of the major brain regions and blood-brain-barrier (BBB) which are similar to a human structure. A few papers described in this review, using genetic and xenotransplantation zebrafish models have been used to study GBM as well as to test the anti-tumoral efficacy of new drugs, their ability to interact with target cells, modulate the tumor microenvironment, cross the BBB and/or their toxicity. Prospective studies following these lines of research may lead to a better diagnosis, prognosis and treatment of patients with GBM.
Collapse
Affiliation(s)
- Pedro Reimunde
- Department of Medicine, Campus de Oza, Universidade da Coruña, 15006 A Coruña, Spain
- Department of Neurosurgery, Hospital Universitario Lucus Augusti, 27003 Lugo, Spain
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Martín Carreira Crende
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Vanesa Lombao Iglesias
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus de Lugo, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.P.-L.); (M.C.C.); (V.L.I.); (L.S.)
| | - Marta Torrecilla-Parra
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Cristina M. Ramírez
- IMDEA Research Institute of Food and Health Sciences, 28049 Madrid, Spain; (M.T.-P.); (C.M.R.)
| | - Clément Anfray
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| | - Fernando Torres Andón
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Universidade de Santiago de Compostela, 15706 Santiago de Compostela, Spain
- IRCCS Istituto Clinico Humanitas, Via A. Manzoni 56, 20089 Rozzano, Milan, Italy;
| |
Collapse
|