1
|
Liu T, Shang D, Tian W, Li Y, Xie R, Zhao J, Dong H. The concentration of dissolved organic matter impacts the neurobehavior in zebrafish larvae exposed to cyclophosphamide. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:61181-61190. [PMID: 39404950 DOI: 10.1007/s11356-024-34907-1] [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: 01/17/2024] [Accepted: 08/27/2024] [Indexed: 11/05/2024]
Abstract
Dissolved organic matter (DOM) occurs ubiquitously in various water matrices and affects the chemical speciation and toxicity of emerging contaminants, such as cyclophosphamide (CP). However, the effects of CP in aquatic organisms with the presence of DOM have been relatively less addressed. In this study, zebrafish eggs < 4 h post fertilization (hpf) were exposed to CP (0 and 50 μg/L) and humic acid (HA, a main component of DOM, 0, 3, 10, and 30 mg-C/L) until 7 days post fertilization, and its toxicity was evaluated by behavioral approaches and transcription of nervous-related genes. An increase in swimming velocity and anxiety was noticed in zebrafish larvae exposed to CP. The related genes of neurotransmitter (drd1, mao, thp1b, and gad2), neurodevelopment (gli2b, nrd, and gfap), and neuroinflammation (thfα, casp3, and il-6) were upregulated by CP. In the presence of HA (3 mg-C/L), the behaviors and gene transcripts of zebrafish larvae were enhanced, while at 10 mg-C/L, they were mitigated. This study has demonstrated that DOM at low concentration increases the toxicity of CP and at high concentration alleviates its toxicity. This study highlights the importance of emerging contaminant exposure with the presence of DOM on their toxicities in aquatic organisms.
Collapse
Affiliation(s)
- Tianming Liu
- China MCC20 Group Co., Ltd, Pangu Road 777, Shanghai, 325035, China.
| | - Dongshen Shang
- China MCC20 Group Co., Ltd, Pangu Road 777, Shanghai, 325035, China
| | - Weiqi Tian
- China MCC20 Group Co., Ltd, Pangu Road 777, Shanghai, 325035, China
| | - Yindong Li
- China MCC20 Group Co., Ltd, Pangu Road 777, Shanghai, 325035, China
| | - Rong Xie
- China MCC20 Group Co., Ltd, Pangu Road 777, Shanghai, 325035, China
| | - Jianxing Zhao
- China MCC20 Group Co., Ltd, Pangu Road 777, Shanghai, 325035, China
| | - Heru Dong
- China MCC20 Group Co., Ltd, Pangu Road 777, Shanghai, 325035, China
| |
Collapse
|
2
|
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
|
3
|
Arora L, Patra D, Roy S, Nanda S, Singh N, Verma AK, Chakraborti A, Dasgupta S, Pal D. Hypoxia-induced miR-210-3p expression in lung adenocarcinoma potentiates tumor development by regulating CCL2 mediated monocyte infiltration. Mol Oncol 2024; 18:1278-1300. [PMID: 35658112 PMCID: PMC11077004 DOI: 10.1002/1878-0261.13260] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/24/2022] [Accepted: 06/02/2022] [Indexed: 11/09/2022] Open
Abstract
In most cancers, tumor hypoxia downregulates the expression of C-C motif chemokine 2 (CCL2), and this downregulation has been implicated in monocyte infiltration and tumor progression; however, the molecular mechanism is not yet clear. We compared noncancerous and lung-adenocarcinoma human samples for hypoxia-inducible factor 1-alpha (HIF-1A), microRNA-210-3p (mir-210-3p), and CCL2 levels. Mechanistic studies were performed on lung adenocarcinoma cell lines and 3D tumor spheroids to understand the role of hypoxia-induced miR-210-3p in the regulation of CCL2 expression and macrophage polarization. HIF-1Α stabilization increases miR-210-3p levels in lung adenocarcinoma and impairs monocyte infiltration by inhibiting CCL2 expression. Mechanistically, miR-210-3p directly binds to the 3'untranslated region (UTR) of CCL2 mRNA and silences it. Suppressing miR-210-3p substantially downregulates the effect of hypoxia on CCL2 expression. Monocyte migration is significantly hampered in miR-210-3p mimic-transfected HIF-1A silenced cancer cells. In contrast, inhibition of miR-210-3p in HIF-1A-overexpressed cells markedly restored monocyte migration, highlighting a direct link between the miR-210-3p level and tumor monocyte burden. Moreover, miR-210-3p inhibition in 3D tumor spheroids promotes monocyte recruitment and skewing towards an antitumor M1 phenotype. Anti-hsa-miR-210-3p-locked nucleic acid (LNA) delivery in a lung tumor xenograft zebrafish model caused tumor regression, suggesting that miR-210-3p could be a promising target for immunomodulatory therapeutic strategies against lung adenocarcinoma.
Collapse
Affiliation(s)
- Leena Arora
- Department of Biomedical EngineeringIndian Institute of Technology RoparPunjabIndia
| | - Debarun Patra
- Department of Biomedical EngineeringIndian Institute of Technology RoparPunjabIndia
| | - Soumyajit Roy
- Department of Biomedical EngineeringIndian Institute of Technology RoparPunjabIndia
| | - Sidhanta Nanda
- Department of Biomedical EngineeringIndian Institute of Technology RoparPunjabIndia
| | - Navneet Singh
- Department of Pulmonary MedicinePostgraduate Institute of Medical Education & Research (PGIMER)ChandigarhIndia
| | - Anita K. Verma
- Department of Zoology, Kirori Mal CollegeUniversity of DelhiIndia
| | - Anuradha Chakraborti
- Department of Experimental Medicine & BiotechnologyPostgraduate Institute of Medical Education & Research (PGIMER)ChandigarhIndia
| | - Suman Dasgupta
- Department of Molecular Biology & BiotechnologyTezpur UniversityAssamIndia
| | - Durba Pal
- Department of Biomedical EngineeringIndian Institute of Technology RoparPunjabIndia
| |
Collapse
|
4
|
Xiao B, Landesman-Bollag E, Feng H. What value do zebrafish have to anticancer drug discovery? Expert Opin Drug Discov 2024; 19:369-375. [PMID: 38327017 PMCID: PMC10950524 DOI: 10.1080/17460441.2024.2313454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Affiliation(s)
- Boyuan Xiao
- Department of Pharmacology, Physiology & Biophysics, Cancer Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, USA
| | - Esther Landesman-Bollag
- Department of Medicine, Section of Hematology and Medical Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, USA
| | - Hui Feng
- Department of Pharmacology, Physiology & Biophysics, Cancer Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, USA
- Department of Medicine, Section of Hematology and Medical Oncology, Boston University Chobanian & Avedisian School of Medicine, Boston, USA
| |
Collapse
|
5
|
Thomas JR, Frye WJE, Robey RW, Gottesman MM. Progress in characterizing ABC multidrug transporters in zebrafish. Drug Resist Updat 2024; 72:101035. [PMID: 38141369 PMCID: PMC10843779 DOI: 10.1016/j.drup.2023.101035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 12/25/2023]
Abstract
Zebrafish have proved to be invaluable for modeling complex physiological processes shared by all vertebrate animals. Resistance of cancers and other diseases to drug treatment can occur owing to expression of the ATP-dependent multidrug transporters ABCB1, ABCG2, and ABCC1, either because of expression of these transporters by the target cells to reduce intracellular concentrations of cytotoxic drugs at barrier sites such as the blood-brain barrier (BBB) to limit penetration of drugs into privileged compartments, or by affecting the absorption, distribution, and excretion of drugs administered orally, through the skin, or directly into the bloodstream. We describe the drug specificity, cellular localization, and function of zebrafish orthologs of multidrug resistance ABC transporters with the goal of developing zebrafish models to explore the physiological and pathophysiological functions of these transporters. Finally, we provide context demonstrating the utility of zebrafish in studying cancer drug resistance. Our ultimate goal is to improve treatment of cancer and other diseases which are affected by ABC multidrug resistance transporters.
Collapse
Affiliation(s)
- Joanna R Thomas
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William J E Frye
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Robert W Robey
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
6
|
Wang W, Li Y, Lin K, Wang X, Tu Y, Zhuo Z. Progress in building clinically relevant patient-derived tumor xenograft models for cancer research. Animal Model Exp Med 2023; 6:381-398. [PMID: 37679891 PMCID: PMC10614132 DOI: 10.1002/ame2.12349] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023] Open
Abstract
Patient-derived tumor xenograft (PDX) models, a method involving the surgical extraction of tumor tissues from cancer patients and subsequent transplantation into immunodeficient mice, have emerged as a pivotal approach in translational research, particularly in advancing precision medicine. As the first stage of PDX development, the patient-derived orthotopic xenograft (PDOX) models implant tumor tissue in mice in the corresponding anatomical locations of the patient. The PDOX models have several advantages, including high fidelity to the original tumor, heightened drug sensitivity, and an elevated rate of successful transplantation. However, the PDOX models present significant challenges, requiring advanced surgical techniques and resource-intensive imaging technologies, which limit its application. And then, the humanized mouse models, as well as the zebrafish models, were developed. Humanized mouse models contain a human immune environment resembling the tumor and immune system interplay. The humanized mouse models are a hot topic in PDX model research. Regarding zebrafish patient-derived tumor xenografts (zPDX) and patient-derived organoids (PDO) as promising models for studying cancer and drug discovery, zPDX models are used to transplant tumors into zebrafish as novel personalized medical animal models with the advantage of reducing patient waiting time. PDO models provide a cost-effective approach for drug testing that replicates the in vivo environment and preserves important tumor-related information for patients. The present review highlights the functional characteristics of each new phase of PDX and provides insights into the challenges and prospective developments in this rapidly evolving field.
Collapse
Affiliation(s)
- Weijing Wang
- Department of Clinical MedicineShantou University Medical CollegeShantouChina
| | - Yongshu Li
- College of Life SciencesHubei Normal UniversityHuangshiChina
- Shenzhen Institute for Technology InnovationNational Institute of MetrologyShenzhenChina
| | - Kaida Lin
- Department of Clinical MedicineShantou University Medical CollegeShantouChina
| | - Xiaokang Wang
- Department of PharmacyShenzhen Longhua District Central HospitalShenzhenChina
| | - Yanyang Tu
- Research Center, Huizhou Central People's HospitalGuangdong Medical UniversityHuizhou CityChina
| | - Zhenjian Zhuo
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
- Laboratory Animal Center, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| |
Collapse
|
7
|
Cao XZ, Zhang BQ, Wang CF, Yin JN, Haider W, Said G, Wei MY, Lu L. A Terphenyllin Derivative CHNQD-00824 from the Marine Compound Library Induced DNA Damage as a Potential Anticancer Agent. Mar Drugs 2023; 21:512. [PMID: 37888447 PMCID: PMC10608154 DOI: 10.3390/md21100512] [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: 09/12/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/28/2023] Open
Abstract
With the emergence of drug resistance and the consequential high morbidity and mortality rates, there is an urgent need to screen and identify new agents for the effective treatment of cancer. Terphenyls-a group of aromatic hydrocarbons consisting of a linear 1,4-diaryl-substituted benzene core-has exhibited a wide range of biological activities. In this study, we discovered a terphenyllin derivative-CHNQD-00824-derived from the marine compound library as a potential anticancer agent. The cytotoxic activities of the CHNQD-00824 compound were evaluated against 13 different cell lines with IC50 values from 0.16 to 7.64 μM. Further study showed that CHNQD-00824 inhibited the proliferation and migration of cancer cells, possibly by inducing DNA damage. Acridine orange staining demonstrated that CHNQD-00824 promoted apoptosis in zebrafish embryos. Notably, the anti-cancer effectiveness was verified in a doxycin hydrochloride (DOX)-induced liver-specific enlargement model in zebrafish. With Solafinib as a positive control, CHNQD-00824 markedly suppressed tumor growth at concentrations of 2.5 and 5 μM, further highlighting its potential as an effective anticancer agent.
Collapse
Affiliation(s)
- Xi-Zhen Cao
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Bo-Qi Zhang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Cui-Fang Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Jun-Na Yin
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Waqas Haider
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Gulab Said
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Department of Chemistry, Women University Swabi, Swabi 23430, Pakistan
| | - Mei-Yan Wei
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| | - Ling Lu
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (X.-Z.C.); (B.-Q.Z.); (C.-F.W.); (J.-N.Y.); (W.H.); (G.S.); (M.-Y.W.)
- Laboratory for Marine Drugs and Biological Products, Laoshan Laboratory, Qingdao 266003, China
| |
Collapse
|
8
|
Hedge JM, Hunter DL, Sanders E, Jarema KA, Olin JK, Britton KN, Lowery M, Knapp BR, Padilla S, Hill BN. Influence of Methylene Blue or Dimethyl Sulfoxide on Larval Zebrafish Development and Behavior. Zebrafish 2023; 20:132-145. [PMID: 37406269 PMCID: PMC10627343 DOI: 10.1089/zeb.2023.0017] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023] Open
Abstract
The use of larval zebrafish developmental testing and assessment, specifically larval zebrafish locomotor activity, has been recognized as a higher throughput testing strategy to identify developmentally toxic and neurotoxic chemicals. There are, however, no standardized protocols for this type of assay, which could result in confounding variables being overlooked. Two chemicals commonly employed during early-life stage zebrafish assays, methylene blue (antifungal agent) and dimethyl sulfoxide (DMSO, a commonly used vehicle) have been reported to affect the morphology and behavior of freshwater fish. In this study, we conducted developmental toxicity (morphology) and neurotoxicity (behavior) assessments of commonly employed concentrations for both chemicals (0.6-10.0 μM methylene blue; 0.3%-1.0% v/v DMSO). A light-dark transition behavioral testing paradigm was applied to morphologically normal, 6 days postfertilization (dpf) zebrafish larvae kept at 26°C. Additionally, an acute DMSO challenge was administered based on early-life stage zebrafish assays typically used in this research area. Results from developmental toxicity screens were similar between both chemicals with no morphological abnormalities detected at any of the concentrations tested. However, neurodevelopmental results were mixed between the two chemicals of interest. Methylene blue resulted in no behavioral changes up to the highest concentration tested, 10.0 μM. By contrast, DMSO altered larval behavior following developmental exposure at concentrations as low as 0.5% (v/v) and exhibited differential concentration-response patterns in the light and dark photoperiods. These results indicate that developmental DMSO exposure can affect larval zebrafish locomotor activity at routinely used concentrations in developmental neurotoxicity assessments, whereas methylene blue does not appear to be developmentally or neurodevelopmentally toxic to larval zebrafish at routinely used concentrations. These results also highlight the importance of understanding the influence of experimental conditions on larval zebrafish locomotor activity that may ultimately confound the interpretation of results.
Collapse
Affiliation(s)
- Joan M. Hedge
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Advanced Experimental Toxicology Models Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Deborah L. Hunter
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Erik Sanders
- Aquatics Lab Services LLC 1112 Nashville Street St. Peters, MO 63376, USA
| | - Kimberly A. Jarema
- Office of Research and Development, Center for Public Health and Environmental Assessment, Immediate Office, Program Operations Staff, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Jeanene K. Olin
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Katy N. Britton
- ORAU Research Participation Program hosted by EPA, Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Morgan Lowery
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Bridget R. Knapp
- ORISE Research Participation Program hosted by EPA, Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Stephanie Padilla
- Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| | - Bridgett N. Hill
- ORISE Research Participation Program hosted by EPA, Office of Research and Development, Center for Computational Toxicology and Exposure, Biomolecular and Computational Toxicology Division, Rapid Assay Development Branch, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, USA
| |
Collapse
|
9
|
Yin J, Zhao G, Kalirai H, Coupland SE, Jochemsen AG, Forn-Cuní G, Wierenga APA, Jager MJ, Snaar-Jagalska BE, Groenewoud A. Zebrafish Patient-Derived Xenograft Model as a Preclinical Platform for Uveal Melanoma Drug Discovery. Pharmaceuticals (Basel) 2023; 16:598. [PMID: 37111355 PMCID: PMC10141637 DOI: 10.3390/ph16040598] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/06/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Uveal melanoma (UM) is a rare malignant cancer of the eye, with up to 50% of patients dying from metastasis, for which no effective treatment is available. Due to the rarity of the disease, there is a great need to harness the limited material available from primary tumors and metastases for advanced research and preclinical drug screening. We established a platform to isolate, preserve, and transiently recover viable tissues, followed by the generation of spheroid cultures derived from primary UM. All assessed tumor-derived samples formed spheroids in culture within 24 h and stained positive for melanocyte-specific markers, indicating the retention of their melanocytic origin. These short-lived spheroids were only maintained for the duration of the experiment (7 days) or re-established from frozen tumor tissue acquired from the same patient. Intravenous injection of fluorescently labeled UM cells derived from these spheroids into zebrafish yielded a reproducible metastatic phenotype and recapitulated molecular features of the disseminating UM. This approach allowed for the experimental replications required for reliable drug screening (at least 2 individual biological experiments, with n > 20). Drug treatments with navitoclax and everolimus validated the zebrafish patient-derived model as a versatile preclinical tool for screening anti-UM drugs and as a preclinical platform to predict personalized drug responses.
Collapse
Affiliation(s)
- Jie Yin
- Institute of Biology, Leiden University, 2333 BE Leiden, The Netherlands; (J.Y.)
| | - Gangyin Zhao
- Institute of Biology, Leiden University, 2333 BE Leiden, The Netherlands; (J.Y.)
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Sarah E. Coupland
- Liverpool Ocular Oncology Research Centre, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool L69 3BX, UK
| | - Aart G. Jochemsen
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Gabriel Forn-Cuní
- Institute of Biology, Leiden University, 2333 BE Leiden, The Netherlands; (J.Y.)
| | - Annemijn P. A. Wierenga
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Arwin Groenewoud
- Institute of Biology, Leiden University, 2333 BE Leiden, The Netherlands; (J.Y.)
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| |
Collapse
|
10
|
Tamburello M, Abate A, Rossini E, Basnet RM, Zizioli D, Cosentini D, Hantel C, Laganà M, Tiberio GAM, Grisanti S, Memo M, Berruti A, Sigala S. Preclinical Evidence of Progesterone as a New Pharmacological Strategy in Human Adrenocortical Carcinoma Cell Lines. Int J Mol Sci 2023; 24:ijms24076829. [PMID: 37047801 PMCID: PMC10095539 DOI: 10.3390/ijms24076829] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023] Open
Abstract
Background: Adrenocortical cancer (ACC) is a rare malignancy with a dismal prognosis. The treatment includes mitotane and EDP chemotherapy (etoposide, doxorubicin, and cisplatin). However, new therapeutic approaches for advanced ACC are needed, particularly targeting the metastatic process. Here, we deepen the role of progesterone as a new potential drug for ACC, in line with its antitumoral effect in other cancers. Methods: NCI-H295R, MUC-1, and TVBF-7 cell lines were used and xenografted in zebrafish embryos. Migration and invasion were studied using transwell assays, and MMP2 activity was studied using zymography. Apoptosis and cell cycle were analyzed by flow cytometry. Results: Progesterone significantly reduced xenograft tumor area and metastasis formation in embryos injected with metastatic lines, MUC-1 and TVBF-7. These results were confirmed in vitro, where the reduction of invasion was mediated, at least in part, by the decrease in MMP2 levels. Progesterone exerted a long-lasting effect in metastaticcells. Progesterone caused apoptosis in NCI-H295R and MUC-1, inducing changes in the cell-cycle distribution, while autophagy was predominantly activated in TVBF-7 cells. Conclusion: Our results give support to the role of progesterone in ACC. The involvement of its analog (megestrol acetate) in reducing ACC progression in ACC patients undergoing EDP-M therapy is now under investigation in the PESETA phase II clinical study.
Collapse
Affiliation(s)
- Mariangela Tamburello
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Andrea Abate
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Elisa Rossini
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Ram Manohar Basnet
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Daniela Zizioli
- Section of Biotechnology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Deborah Cosentini
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Constanze Hantel
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University of Zurich (UZH), 8091 Zürich, Switzerland
- Medizinische Klinik und Poliklinik III, University Hospital Carl Gustav Carus Dresden, 01307 Dresden, Germany
| | - Marta Laganà
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Guido Alberto Massimo Tiberio
- Surgical Clinic, Department of Clinical and Experimental Sciences, University of Brescia at ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Salvatore Grisanti
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Maurizio Memo
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| | - Alfredo Berruti
- Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Sandra Sigala
- Section of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy
| |
Collapse
|
11
|
Morris EK, Daignault-Mill S, Stehbens SJ, Genovesi LA, Lagendijk AK. Addressing blood-brain-tumor-barrier heterogeneity in pediatric brain tumors with innovative preclinical models. Front Oncol 2023; 13:1101522. [PMID: 36776301 PMCID: PMC9909546 DOI: 10.3389/fonc.2023.1101522] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/06/2023] [Indexed: 01/27/2023] Open
Abstract
Brain tumors represent the leading cause of disease-related mortality and morbidity in children, with effective treatments urgently required. One factor limiting the effectiveness of systemic therapy is the blood-brain-barrier (BBB), which limits the brain penetration of many anticancer drugs. BBB integrity is often compromised in tumors, referred to as the blood-brain-tumor-barrier (BBTB), and the impact of a compromised BBTB on the therapeutic sensitivity of brain tumors has been clearly shown for a few selected agents. However, the heterogeneity of barrier alteration observed within a single tumor and across distinct pediatric tumor types represents an additional challenge. Herein, we discuss what is known regarding the heterogeneity of tumor-associated vasculature in pediatric brain tumors. We discuss innovative and complementary preclinical model systems that will facilitate real-time functional analyses of BBTB for all pediatric brain tumor types. We believe a broader use of these preclinical models will enable us to develop a greater understanding of the processes underlying tumor-associated vasculature formation and ultimately more efficacious treatment options.
Collapse
Affiliation(s)
- Elysse K. Morris
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Sheena Daignault-Mill
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Samantha J. Stehbens
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia
| | - Laura A. Genovesi
- The University of Queensland Frazer Institute, Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia,*Correspondence: Laura A. Genovesi, ; Anne K. Lagendijk,
| | - Anne K. Lagendijk
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, Australia,School of Biomedical Sciences, University of Queensland, St. Lucia, QLD, Australia,*Correspondence: Laura A. Genovesi, ; Anne K. Lagendijk,
| |
Collapse
|
12
|
Gatzweiler C, Ridinger J, Ayhan S, Najafi S, Peterziel H, Witt O, Oehme I. Evaluation of Antitumor and On-Target Activity of HDAC Inhibitors with the Zebrafish Embryo Xenograft Model. Methods Mol Biol 2023; 2589:75-85. [PMID: 36255618 DOI: 10.1007/978-1-0716-2788-4_5] [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] [Indexed: 06/16/2023]
Abstract
Reliable preclinical drug testing models for cancer research are urgently needed with zebrafish embryo models emerging as a powerful vertebrate model for xenotransplantation studies. Here, we describe the evaluation of toxicity, efficacy, and on-target activity of histone deacetylase (HDAC) inhibitors in a zebrafish embryo yolk sac xenotransplantation model of medulloblastoma and neuroblastoma cells. For this, we performed toxicity assays with our zebrafish drug library consisting of 28 clinically relevant targeted as well as chemotherapeutic drugs with zebrafish embryos. We further engrafted zebrafish embryos with fluorescently labeled pediatric tumor cells (SK-N-BE(2)-C, HD-MB03, or MED8A) and monitored the progression after HDAC inhibitor treatment of xenotransplanted tumors through tumor volume measurements with high-content confocal microscopy in a multi-well format. The on-target activity of HDAC inhibitors was verified through immunohistochemistry staining on paraffin-embedded early larvae. Overall, the zebrafish embryo xenotransplantation model allows for fast and cost-efficient in vivo evaluation of targeted drug toxicity, efficacy, and on-target activity in the field of precision oncology.
Collapse
Affiliation(s)
- Charlotte Gatzweiler
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Johannes Ridinger
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simay Ayhan
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Sara Najafi
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Heike Peterziel
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Ina Oehme
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany.
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
13
|
Li X, Li M. The application of zebrafish patient-derived xenograft tumor models in the development of antitumor agents. Med Res Rev 2023; 43:212-236. [PMID: 36029178 DOI: 10.1002/med.21924] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/09/2022] [Accepted: 07/28/2022] [Indexed: 02/04/2023]
Abstract
The cost of antitumor drug development is enormous, yet the clinical outcomes are less than satisfactory. Therefore, it is of great importance to develop effective drug screening methods that enable accurate, rapid, and high-throughput discovery of lead compounds in the process of preclinical antitumor drug research. An effective solution is to use the patient-derived xenograft (PDX) tumor animal models, which are applicable for the elucidation of tumor pathogenesis and the preclinical testing of novel antitumor compounds. As a promising screening model organism, zebrafish has been widely applied in the construction of the PDX tumor model and the discovery of antineoplastic agents. Herein, we systematically survey the recent cutting-edge advances in zebrafish PDX models (zPDX) for studies of pathogenesis mechanisms and drug screening. In addition, the techniques used in the construction of zPDX are summarized. The advantages and limitations of the zPDX are also discussed in detail. Finally, the prospects of zPDX in drug discovery, translational medicine, and clinical precision medicine treatment are well presented.
Collapse
Affiliation(s)
- Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Minyong Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
14
|
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: 8] [Impact Index Per Article: 4.0] [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
|
15
|
Yi ZN, Chen XK, Ma ACH. Modeling leukemia with zebrafish (Danio rerio): Towards precision medicine. Exp Cell Res 2022; 421:113401. [PMID: 36306826 DOI: 10.1016/j.yexcr.2022.113401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 12/29/2022]
Abstract
Leukemia is a type of blood cancer characterized by high genetic heterogeneity and fatality. While chemotherapy remains the primary form of treatment for leukemia, its effectiveness was profoundly diminished by the genetic heterogeneity and cytogenetic abnormalities of leukemic cells. Therefore, there is an unmet need to develop precision medicine for leukemia with distinct genetic backgrounds. Zebrafish (Danio rerio), a freshwater fish with exceptional feasibility in genome editing, is a powerful tool for rapid human cancer modeling. In the past decades, zebrafish have been adopted in modeling human leukemia, exploring the molecular mechanisms of underlying genetic abnormalities, and discovering novel therapeutic agents. Although many recurrent mutations of leukemia have been modeled in zebrafish for pathological study and drug discovery, its great potential in leukemia modeling was not yet fully exploited, particularly in precision medicine. In this review, we evaluated the current zebrafish models of leukemia/pre-leukemia and genetic techniques and discussed the potential of zebrafish models with novel techniques, which may contribute to the development of zebrafish as a disease model for precision medicine in treating leukemia.
Collapse
Affiliation(s)
- Zhen-Ni Yi
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xiang-Ke Chen
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Alvin Chun-Hang Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China.
| |
Collapse
|
16
|
Ganesan R, Mukherjee AG, Gopalakrishnan AV, Prabhakaran VS. Solid-State NMR-Based Metabolomics Imprinting Elucidation in Tissue Metabolites, Metabolites Inhibition, and Metabolic Hub in Zebrafish by Chitosan. Metabolites 2022; 12:metabo12121263. [PMID: 36557301 PMCID: PMC9785866 DOI: 10.3390/metabo12121263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
In this study, we demonstrated that chitosan-applied zebrafish (Danio rerio) tissue metabolite alteration, metabolic discrimination, and metabolic phenotypic expression occurred. The spectroscopy of solid-state 1H nuclear magnetic resonance (ss 1H-NMR) has been used. Chitosan has no, or low, toxicity and is a biocompatible biomaterial; however, the metabolite mechanisms underlying the biological effect of chitosan are poorly understood. The zebrafish is now one of the most popular ecotoxicology models. Zebrafish were exposed to chitosan concentrations of 0, 50, 100, 200, and 500 mg/L, and the body tissue was subjected to metabolites-targeted profiling. The zebrafish samples were measured via solvent-suppressed and T2-filtered methods with in vivo zebrafish metabolites. The metabolism of glutamate, glutamine, glutathione (GSH), taurine, trimethylamine (TMA), and its N-oxide (TMAO) is also significantly altered. Here, we report the quantification of metabolites and the biological application of chitosan. The metabolomics profile of chitosan in zebrafish has been detected, and the results indicated disturbed amino acid metabolism, the TCA cycle, and glycolysis. Our results demonstrate the potential of comparative metabolite profiling for discovering bioactive metabolites and they highlight the power of chitosan-applied chemical metabolomics to uncover new biological insights.
Collapse
Affiliation(s)
- Raja Ganesan
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24253, Republic of Korea
- Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
- Correspondence: (R.G.); (A.V.G.)
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
- Correspondence: (R.G.); (A.V.G.)
| | - Vasantha-Srinivasan Prabhakaran
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, India
| |
Collapse
|
17
|
Cho HJ, Lee WS, Jeong J, Lee JS. A review on the impacts of nanomaterials on neuromodulation and neurological dysfunction using a zebrafish animal model. Comp Biochem Physiol C Toxicol Pharmacol 2022; 261:109428. [PMID: 35940544 DOI: 10.1016/j.cbpc.2022.109428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 11/20/2022]
Abstract
Nanomaterials have been widely employed from industrial to medical fields due to their small sizes and versatile characteristics. However, nanomaterials can also induce unexpected adverse effects on health. In particular, exposure of the nervous system to nanomaterials can cause serious neurological dysfunctions and neurodegenerative diseases. A number of studies have adopted various animal models to evaluate the neurotoxic effects of nanomaterials. Among them, zebrafish has become an attractive animal model for neurotoxicological studies due to several advantages, including the well-characterized nervous system, efficient genome editing, convenient generation of transgenic lines, high-resolution in vivo imaging, and an array of behavioral assays. In this review, we summarize recent studies on the neurotoxicological effects of nanomaterials, particularly engineered nanomaterials and nanoplastics, using zebrafish and discuss key findings with advantages and limitations of the zebrafish model in neurotoxicological studies.
Collapse
Affiliation(s)
- Hyun-Ju Cho
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Wang Sik Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jinyoung Jeong
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; KRIBB School, University of Science and Technology, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| | - Jeong-Soo Lee
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea; KRIBB School, University of Science and Technology, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
18
|
Yang G, Wang L, Qin X, Chen X, Liang Y, Jin X, Chen C, Zhang W, Pan W, Li H. Heterogeneities of zebrafish vasculature development studied by a high throughput light-sheet flow imaging system. BIOMEDICAL OPTICS EXPRESS 2022; 13:5344-5357. [PMID: 36425637 PMCID: PMC9664872 DOI: 10.1364/boe.470058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/17/2022] [Accepted: 08/30/2022] [Indexed: 06/16/2023]
Abstract
Zebrafish is one of the ideal model animals to study the structural and functional heterogeneities in development. However, the lack of high throughput 3D imaging techniques has limited studies to only a few samples, despite zebrafish spawning tens of embryos at once. Here, we report a light-sheet flow imaging system (LS-FIS) based on light-sheet illumination and a continuous flow imager. LS-FIS enables whole-larva 3D imaging of tens of samples within half an hour. The high throughput 3D imaging capability of LS-FIS was demonstrated with the developmental study of the zebrafish vasculature from 3 to 9 days post-fertilization. Statistical analysis shows significant variances in trunk vessel development but less in hyaloid vessel development.
Collapse
Affiliation(s)
- Guang Yang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou 215163, China
- Jiangsu Key Laboratory of Medical Optics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Linbo Wang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou 215163, China
- Jiangsu Key Laboratory of Medical Optics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Xiaofei Qin
- Jiangsu Key Laboratory of Medical Optics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Xiaohu Chen
- Jiangsu Key Laboratory of Medical Optics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Yong Liang
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou 215163, China
- Jiangsu Key Laboratory of Medical Optics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Xin Jin
- Jiangsu Key Laboratory of Medical Optics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Chong Chen
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou 215163, China
- Jiangsu Key Laboratory of Medical Optics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| | - Wenjuan Zhang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weijun Pan
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Li
- Jiangsu Key Laboratory of Medical Optics,
Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China
| |
Collapse
|
19
|
Bozzer S, De Maso L, Grimaldi MC, Capolla S, Dal Bo M, Toffoli G, Macor P. Zebrafish: A Useful Animal Model for the Characterization of Drug-Loaded Polymeric NPs. Biomedicines 2022; 10:biomedicines10092252. [PMID: 36140353 PMCID: PMC9496256 DOI: 10.3390/biomedicines10092252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 11/28/2022] Open
Abstract
The use of zebrafish (ZF) embryos as an in vivo model is increasingly attractive thanks to different features that include easy handling, transparency, and the absence of adaptive immunity until 4–6 weeks. These factors allow the development of xenografts that can be easily analyzed through fluorescence techniques. In this work, ZF were exploited to characterize the efficiency of drug-loaded polymeric NPs as a therapeutical approach for B-cell malignancies. Fluorescent probes, fluorescent transgenic lines of ZF, or their combination allowed to deeply examine biodistribution, elimination, and therapeutic efficacy. In particular, the fluorescent signal of nanoparticles (NPs) was exploited to investigate the in vivo distribution, while the colocalization between the fluorescence in macrophages and NPs allows following the elimination pathway of these polymeric NPs. Xenotransplanted human B-cells (Nalm-6) developed a reproducible model useful for demonstrating drug delivery by polymeric NPs loaded with doxorubicin and, as a consequence, the arrest of tumor growth and the reduction in tumor burden. ZF proved to be a versatile model, able to rapidly provide answers in the development of animal models and in the characterization of the activity and the efficacy of drug delivery systems.
Collapse
Affiliation(s)
- Sara Bozzer
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- Correspondence: (S.B.); (P.M.)
| | - Luca De Maso
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | | | - Sara Capolla
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- Correspondence: (S.B.); (P.M.)
| |
Collapse
|
20
|
Dudziak K, Nowak M, Sozoniuk M. One Host-Multiple Applications: Zebrafish ( Danio rerio) as Promising Model for Studying Human Cancers and Pathogenic Diseases. Int J Mol Sci 2022; 23:10255. [PMID: 36142160 PMCID: PMC9499349 DOI: 10.3390/ijms231810255] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/03/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, zebrafish (ZF) has been increasingly applied as a model in human disease studies, with a particular focus on cancer. A number of advantages make it an attractive alternative for mice widely used so far. Due to the many advantages of zebrafish, modifications can be based on different mechanisms and the induction of human disease can take different forms depending on the research goal. Genetic manipulation, tumor transplantation, or injection of the pathogen are only a few examples of using ZF as a model. Most of the studies are conducted in order to understand the disease mechanism, monitor disease progression, test new or alternative therapies, and select the best treatment. The transplantation of cancer cells derived from patients enables the development of personalized medicine. To better mimic a patient's body environment, immune-deficient models (SCID) have been developed. A lower immune response is mostly generated by genetic manipulation but also by irradiation or dexamethasone treatment. For many studies, using SCID provides a better chance to avoid cancer cell rejection. In this review, we describe the main directions of using ZF in research, explain why and how zebrafish can be used as a model, what kind of limitations will be met and how to overcome them. We collected recent achievements in this field, indicating promising perspectives for the future.
Collapse
Affiliation(s)
- Karolina Dudziak
- Chair and Department of Biochemistry and Molecular Biology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Michał Nowak
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| | - Magdalena Sozoniuk
- Institute of Plant Genetics, Breeding and Biotechnology, University of Life Sciences in Lublin, 20-950 Lublin, Poland
| |
Collapse
|
21
|
Chang YC, Shieh MC, Chang YH, Huang WL, Su WC, Cheng FY, Cheung CHA. Development of a cancer cells self‑activating and miR‑125a‑5p expressing poly‑pharmacological nanodrug for cancer treatment. Int J Mol Med 2022; 50:102. [PMID: 35703361 PMCID: PMC9239037 DOI: 10.3892/ijmm.2022.5158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer cells can acquire resistance to targeted therapeutic agents when the designated targets or their downstream signaling molecules develop protein conformational or activity changes. There is an increasing interest in developing poly-pharmacologic anticancer agents to target multiple oncoproteins or signaling pathways in cancer cells. The microRNA 125a-5p (miR-125a-5p) is a tumor suppressor, and its expression has frequently been downregulated in tumors. By contrast, the anti-apoptotic molecule BIRC5/SURVIVIN is highly expressed in tumors but not in the differentiated normal tissues. In the present study, the development of a BIRC5 gene promoter-driven, miR-125a-5p expressing, poly-L-lysine-conjugated magnetite iron poly-pharmacologic nanodrug (pL-MNP-pSur-125a) was reported. The cancer cells self-activating property and the anticancer effects of this nanodrug were examined in both the multidrug efflux protein ABCB1/MDR1-expressing/-non-expressing cancer cells in vitro and in vivo. It was demonstrated that pL-MNP-pSur-125a decreased the expression of ERBB2/HER2, HDAC5, BIRC5, and SP1, which are hot therapeutic targets for cancer in vitro. Notably, pL-MNP-pSur-125a also downregulated the expression of TDO2 in the human KB cervical carcinoma cells. PL-MNP-pSur-125a decreased the viability of various BIRC5-expressing cancer cells, regardless of the tissue origin or the expression of ABCB1, but not of the human BIRC5-non-expressing HMEC-1 endothelial cells. In vivo, pL-MNP-pSur-125a exhibited potent antitumor growth effects, but without inducing liver toxicity, in various zebrafish human-ABCB1-expressing and ABCB1-non-expressing tumor xenograft models. In conclusion, pL-MNP-pSur-125a is an easy-to-prepare and a promising poly-pharmacological anticancer nanodrug that has the potential to manage numerous malignancies, particularly for patients with BIRC5/ABCB1-related drug resistance after prolonged chemotherapeutic treatments.
Collapse
Affiliation(s)
- Yung-Chieh Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Min-Chieh Shieh
- Division of General Surgery, Department of Surgery, Ditmanson Medical Foundation Chia‑Yi Christian Hospital, Chiayi 600566, Taiwan, R.O.C
| | - Yen-Hsuan Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Wei-Lun Huang
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Wu-Chou Su
- Center of Applied Nanomedicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| | - Fong-Yu Cheng
- Department of Chemistry, College of Sciences, Chinese Culture University, Taipei 111396, Taiwan, R.O.C
| | - Chun Hei Antonio Cheung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701401, Taiwan, R.O.C
| |
Collapse
|
22
|
Wang Q, Wang W, Pan W, Lv X, Zhang L, Zheng K, Tian F, Xu C. Case Report: Two Patients With EGFR Exon 20 Insertion Mutanted Non-Small Cell Lung Cancer Precision Treatment Using Patient-Derived Xenografts in Zebrafish Embryos. Front Oncol 2022; 12:884798. [PMID: 35936715 PMCID: PMC9348893 DOI: 10.3389/fonc.2022.884798] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/10/2022] [Indexed: 12/25/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR) exon 20 insertion mutations are uncommon EGFR mutations and generally resistant to first- and second-generation EGFR-tyrosine kinase inhibitors (TKIs). In precision oncology, treatment regimens are tested for improving the clinical outcomes. Zebrafish embryo tumor transplant models are used in cancer research. Methods We report two Chinese females who were diagnosed with stage IV lung adenocarcinoma and shown to harbor EGFR exon 20 insertion mutations by next-generation sequencing (NGS). Then, we established lung cancer patient-derived xenografts using a zebrafish model. The tumor cells were isolated from the patient. For case one, tumor cells were collected from lymph node biopsy, while the tumor cells were obtained from the pleural effusion. Zebrafish were inoculated with tumor cells and placed in the culture medium containing the third-generation EGFR-TKI, osimertinib. Fluorescence microscope photographs were used to record the red fluorescence area, which represented the proliferation and migration of tumor cells in the zebrafish. Results Case one was diagnosed with lung adenocarcinoma (cT4N3M1b, stage IVB) and had an EGFR exon 20 mutation (p. N771delinsHH [abundance 14.08%]). Tumor cell proliferation and migration were significantly reduced in the osimertinib group compared with the control group. The patient received first-line osimertinib (160 mg). According to RECIST v1.1, she achieved a partial response. Case two had stage IVA lung adenocarcinoma with a pleural effusion. The pleural effusion sample was selected to obtain tumor cells for injection, and the zebrafish lung cancer model was established. The proliferation of tumor cells in the osimertinib group was significantly reduced compared to the control group. The migration of tumor cells was not significantly reduced compared to the control group. The patient also received first-line osimertinib (160 mg). The lung lesions were stable, but the pleural effusion was poorly controlled. Conclusion Our study demonstrates the applicability of a zebrafish embryos model as an innovative platform to targeted drug testing. More precise methods are needed to select treatment options in the future.
Collapse
Affiliation(s)
- Qian Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Suqian Hospital of Chinese Medicine, Suqian, China
| | - Wenxian Wang
- Department of Medical Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Weiwei Pan
- Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing, China
| | - Xiaojing Lv
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Lei Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Suqian Hospital of Chinese Medicine, Suqian, China
| | - Kaiming Zheng
- Department of Geriatric Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Fang Tian
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
- *Correspondence: Chunwei Xu, ; Fang Tian,
| | - Chunwei Xu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
- *Correspondence: Chunwei Xu, ; Fang Tian,
| |
Collapse
|
23
|
Kwiatkowska I, Hermanowicz JM, Iwinska Z, Kowalczuk K, Iwanowska J, Pawlak D. Zebrafish—An Optimal Model in Experimental Oncology. Molecules 2022; 27:molecules27134223. [PMID: 35807468 PMCID: PMC9268704 DOI: 10.3390/molecules27134223] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/10/2022] [Accepted: 06/28/2022] [Indexed: 02/02/2023] Open
Abstract
A thorough understanding of cancer pathogenesis is a necessary step in the development of more effective and safer therapy. However, due to the complexity of the process and intricate interactions, studying tumor development is an extremely difficult and challenging task. In bringing this issue closer, different scientific models with various advancement levels are helpful. Cell cultures is a system that is too simple and does not allow for multidirectional research. On the other hand, rodent models, although commonly used, are burdened with several limitations. For this reason, new model organisms that will allow for the studying of carcinogenesis stages and factors reliably involved in them are urgently sought after. Danio rerio, an inconspicuous fish endowed with unique features, is gaining in importance in the world of scientific research. Including it in oncological research brings solutions to many challenges afflicting modern medicine. This article aims to illustrate the usefulness of Danio rerio as a model organism which turns out to be a powerful and unique tool for studying the stages of carcinogenesis and solving the hitherto incomprehensible processes that lead to the development of the disease.
Collapse
Affiliation(s)
- Iwona Kwiatkowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
- Correspondence: ; Tel./Fax: +48-8574-856-01
| | - Justyna Magdalena Hermanowicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
- Department of Clinical Pharmacy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland
| | - Zaneta Iwinska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| | - Krystyna Kowalczuk
- Department of Integrated Medical Care, Medical University of Bialystok, ul. M Skłodowskiej-Curie 7A, 15-096 Bialystok, Poland;
| | - Jolanta Iwanowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (J.M.H.); (Z.I.); (J.I.); (D.P.)
| |
Collapse
|
24
|
Disease modeling by efficient genome editing using a near PAM-less base editor in vivo. Nat Commun 2022; 13:3435. [PMID: 35701478 PMCID: PMC9198099 DOI: 10.1038/s41467-022-31172-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 06/08/2022] [Indexed: 12/18/2022] Open
Abstract
Base Editors are emerging as an innovative technology to introduce point mutations in complex genomes. So far, the requirement of an NGG Protospacer Adjacent Motif (PAM) at a suitable position often limits the base editing possibility to model human pathological mutations in animals. Here we show that, using the CBE4max-SpRY variant recognizing nearly all PAM sequences, we could introduce point mutations for the first time in an animal model with high efficiency, thus drastically increasing the base editing possibilities. With this near PAM-less base editor we could simultaneously mutate several genes and we developed a co-selection method to identify the most edited embryos based on a simple visual screening. Finally, we apply our method to create a zebrafish model for melanoma predisposition based on the simultaneous base editing of multiple genes. Altogether, our results considerably expand the Base Editor application to introduce human disease-causing mutations in zebrafish. Base Editors are emerging as an innovative technology to introduce point mutations in complex genomes. Here the authors describe a near PAM-less base editor and its application in zebrafish to efficiently create disease models harbouring specific point mutations.
Collapse
|
25
|
Chen Q, Cuello-Garibo JA, Bretin L, Zhang L, Ramu V, Aydar Y, Batsiun Y, Bronkhorst S, Husiev Y, Beztsinna N, Chen L, Zhou XQ, Schmidt C, Ott I, Jager MJ, Brouwer AM, Snaar-Jagalska BE, Bonnet S. Photosubstitution in a trisheteroleptic ruthenium complex inhibits conjunctival melanoma growth in a zebrafish orthotopic xenograft model. Chem Sci 2022; 13:6899-6919. [PMID: 35774173 PMCID: PMC9200134 DOI: 10.1039/d2sc01646j] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/13/2022] [Indexed: 12/28/2022] Open
Abstract
In vivo data are rare but essential for establishing the clinical potential of ruthenium-based photoactivated chemotherapy (PACT) compounds, a new family of phototherapeutic drugs that are activated via ligand photosubstitution. Here a novel trisheteroleptic ruthenium complex [Ru(dpp)(bpy)(mtmp)](PF6)2 ([2](PF6)2, dpp = 4,7-diphenyl-1,10-phenanthroline, bpy = 2,2′-bipyridine, mtmp = 2-methylthiomethylpyridine) was synthesized and its light-activated anticancer properties were validated in cancer cell monolayers, 3D tumor spheroids, and in embryonic zebrafish cancer models. Upon green light irradiation, the non-toxic mtmp ligand is selectively cleaved off, thereby releasing a phototoxic ruthenium-based photoproduct capable notably of binding to nuclear DNA and triggering DNA damage and apoptosis within 24–48 h. In vitro, fifteen minutes of green light irradiation (21 mW cm−2, 19 J cm−2, 520 nm) were sufficient to generate high phototherapeutic indexes (PI) for this compound in a range of cancer cell lines including lung (A549), prostate (PC3Pro4), conjunctival melanoma (CRMM1, CRMM2, CM2005.1) and uveal melanoma (OMM1, OMM2.5, Mel270) cancer cell lines. The therapeutic potential of [2](PF6)2 was further evaluated in zebrafish embryo ectopic (PC3Pro4) or orthotopic (CRMM1, CRMM2) tumour models. The ectopic model consisted of red fluorescent PC3Pro4-mCherry cells injected intravenously (IV) into zebrafish, that formed perivascular metastatic lesions at the posterior ventral end of caudal hematopoietic tissue (CHT). By contrast, in the orthotopic model, CRMM1- and CRMM2-mCherry cells were injected behind the eye where they developed primary lesions. The maximally-tolerated dose (MTD) of [2](PF6)2 was first determined for three different modes of compound administration: (i) incubating the fish in prodrug-containing water (WA); (ii) injecting the prodrug intravenously (IV) into the fish; or (iii) injecting the prodrug retro-orbitally (RO) into the fish. To test the anticancer efficiency of [2](PF6)2, the embryos were treated 24 h after engraftment at the MTD. Optimally, four consecutive PACT treatments were performed on engrafted embryos using 60 min drug-to-light intervals and 90 min green light irradiation (21 mW cm−2, 114 J cm−2, 520 nm). Most importantly, this PACT protocol was not toxic to the zebrafish. In the ectopic prostate tumour models, where [2](PF6)2 showed the highest photoindex in vitro (PI > 31), the PACT treatment did not significantly diminish the growth of primary lesions, while in both conjunctival melanoma orthotopic tumour models, where [2](PF6)2 showed more modest photoindexes (PI ∼ 9), retro-orbitally administered PACT treatment significantly inhibited growth of the engrafted tumors. Overall, this study represents the first demonstration in zebrafish cancer models of the clinical potential of ruthenium-based PACT, here against conjunctival melanoma. A new tris-heteroleptic photoactivated chemotherapy ruthenium complex induces apoptosis upon green light activation in a zebrafish orthothopic conjunctival melanoma xenograft model.![]()
Collapse
Affiliation(s)
- Quanchi Chen
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing China.,Institute of Biology, Leiden University Leiden The Netherlands +31-71-527-4980
| | - Jordi-Amat Cuello-Garibo
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Ludovic Bretin
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Liyan Zhang
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Vadde Ramu
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Yasmin Aydar
- Institute of Biology, Leiden University Leiden The Netherlands +31-71-527-4980
| | - Yevhen Batsiun
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Sharon Bronkhorst
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Yurii Husiev
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Nataliia Beztsinna
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Lanpeng Chen
- Institute of Biology, Leiden University Leiden The Netherlands +31-71-527-4980
| | - Xue-Quan Zhou
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| | - Claudia Schmidt
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstrasse 55 D-38106 Braunschweig Germany
| | - Ingo Ott
- Institute of Medicinal and Pharmaceutical Chemistry, Technische Universität Braunschweig Beethovenstrasse 55 D-38106 Braunschweig Germany
| | - Martine J Jager
- Department of Ophthalmology, Leiden University Medical Center Leiden The Netherlands
| | - Albert M Brouwer
- Van't Hoff Institute for Molecular Sciences, University of Amsterdam Science Park 904 1098 XH Amsterdam The Netherlands
| | | | - Sylvestre Bonnet
- Leiden Institute of Chemistry, Leiden University P. O. Box 9502 2300 RA Leiden The Netherlands +31-71-527-4260
| |
Collapse
|
26
|
Almstedt E, Rosén E, Gloger M, Stockgard R, Hekmati N, Koltowska K, Krona C, Nelander S. Real-time evaluation of glioblastoma growth in patient-specific zebrafish xenografts. Neuro Oncol 2022; 24:726-738. [PMID: 34919147 PMCID: PMC9071311 DOI: 10.1093/neuonc/noab264] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Patient-derived xenograft (PDX) models of glioblastoma (GBM) are a central tool for neuro-oncology research and drug development, enabling the detection of patient-specific differences in growth, and in vivo drug response. However, existing PDX models are not well suited for large-scale or automated studies. Thus, here, we investigate if a fast zebrafish-based PDX model, supported by longitudinal, AI-driven image analysis, can recapitulate key aspects of glioblastoma growth and enable case-comparative drug testing. METHODS We engrafted 11 GFP-tagged patient-derived GBM IDH wild-type cell cultures (PDCs) into 1-day-old zebrafish embryos, and monitored fish with 96-well live microscopy and convolutional neural network analysis. Using light-sheet imaging of whole embryos, we analyzed further the invasive growth of tumor cells. RESULTS Our pipeline enables automatic and robust longitudinal observation of tumor growth and survival of individual fish. The 11 PDCs expressed growth, invasion and survival heterogeneity, and tumor initiation correlated strongly with matched mouse PDX counterparts (Spearman R = 0.89, p < 0.001). Three PDCs showed a high degree of association between grafted tumor cells and host blood vessels, suggesting a perivascular invasion phenotype. In vivo evaluation of the drug marizomib, currently in clinical trials for GBM, showed an effect on fish survival corresponding to PDC in vitro and in vivo marizomib sensitivity. CONCLUSIONS Zebrafish xenografts of GBM, monitored by AI methods in an automated process, present a scalable alternative to mouse xenograft models for the study of glioblastoma tumor initiation, growth, and invasion, applicable to patient-specific drug evaluation.
Collapse
Affiliation(s)
- Elin Almstedt
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Emil Rosén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Marleen Gloger
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Rebecka Stockgard
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Neda Hekmati
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Katarzyna Koltowska
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Cecilia Krona
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Sven Nelander
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
27
|
Cascallar M, Alijas S, Pensado-López A, Vázquez-Ríos AJ, Sánchez L, Piñeiro R, de la Fuente M. What Zebrafish and Nanotechnology Can Offer for Cancer Treatments in the Age of Personalized Medicine. Cancers (Basel) 2022; 14:cancers14092238. [PMID: 35565373 PMCID: PMC9099873 DOI: 10.3390/cancers14092238] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer causes millions of deaths each year and thus urgently requires the development of new therapeutic strategies. Nanotechnology-based anticancer therapies are a promising approach, with several formulations already approved and in clinical use. The evaluation of these therapies requires efficient in vivo models to study their behavior and interaction with cancer cells, and to optimize their properties to ensure maximum efficacy and safety. In this way, zebrafish is an important candidate due to its high homology with the human genoma, its large offspring, and the ease in developing specific cancer models. The role of zebrafish as a model for anticancer therapy studies has been highly evidenced, allowing researchers not only to perform drug screenings but also to evaluate novel therapies such as immunotherapies and nanotherapies. Beyond that, zebrafish can be used as an “avatar” model for performing patient-derived xenografts for personalized medicine. These characteristics place zebrafish in an attractive position as a role model for evaluating novel therapies for cancer treatment, such as nanomedicine.
Collapse
Affiliation(s)
- María Cascallar
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
- Center for Research in Molecular Medicine & Chronic Diseases (CIMUS), Campus Vida, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Abi Judit Vázquez-Ríos
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- DIVERSA Technologies S.L., 15782 Santiago de Compostela, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Universidade de Santiago de Compostela, Campus de Lugo, 27002 Lugo, Spain; (A.P.-L.); (L.S.)
- Preclinical Animal Models Group, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Roberto Piñeiro
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- Roche-Chus Joint Unit, Translational Medical Oncology Group, Oncomet, Health Research Institute of Santiago de Compostela, Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Group, Health Research Institute of Santiago de Compostela (IDIS), SERGAS, 15706 Santiago de Compostela, Spain; (M.C.); (S.A.); (A.J.V.-R.)
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain;
- DIVERSA Technologies S.L., 15782 Santiago de Compostela, Spain
- Correspondence: ; Tel.: +34-981-955-704
| |
Collapse
|
28
|
Liu K, Chen H, Wang Y, Jiang L, Li Y. Evolving Insights Into the Biological Function and Clinical Significance of Long Noncoding RNA in Glioblastoma. Front Cell Dev Biol 2022; 10:846864. [PMID: 35531099 PMCID: PMC9068894 DOI: 10.3389/fcell.2022.846864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is one of the most prevalent and aggressive cancers worldwide. The overall survival period of GBM patients is only 15 months even with standard combination therapy. The absence of validated biomarkers for early diagnosis mainly accounts for worse clinical outcomes of GBM patients. Thus, there is an urgent requirement to characterize more biomarkers for the early diagnosis of GBM patients. In addition, the detailed molecular basis during GBM pathogenesis and oncogenesis is not fully understood, highlighting that it is of great significance to elucidate the molecular mechanisms of GBM initiation and development. Recently, accumulated pieces of evidence have revealed the central roles of long noncoding RNAs (lncRNAs) in the tumorigenesis and progression of GBM by binding with DNA, RNA, or protein. Targeting those oncogenic lncRNAs in GBM may be promising to develop more effective therapeutics. Furthermore, a better understanding of the biological function and underlying molecular basis of dysregulated lncRNAs in GBM initiation and development will offer new insights into GBM early diagnosis and develop novel treatments for GBM patients. Herein, this review builds on previous studies to summarize the dysregulated lncRNAs in GBM and their unique biological functions during GBM tumorigenesis and progression. In addition, new insights and challenges of lncRNA-based diagnostic and therapeutic potentials for GBM patients were also introduced.
Collapse
Affiliation(s)
- Kun Liu
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Hong Chen
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
| | - Yuanyuan Wang
- Department of Pathology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
| | - Liping Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Kunming, China
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Yi Li, ; Liping Jiang,
| | - Yi Li
- Department of Oncology, 920th Hospital of Joint Logistics Support Force, Teaching Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Yi Li, ; Liping Jiang,
| |
Collapse
|
29
|
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: 4.0] [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
|
30
|
Costa B, Estrada MF, Barroso MT, Fior R. Zebrafish Patient-Derived Avatars from Digestive Cancers for Anti-cancer Therapy Screening. Curr Protoc 2022; 2:e415. [PMID: 35436037 DOI: 10.1002/cpz1.415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Patient-derived xenografts (PDXs), also called "avatars," are generated by the implantation of human primary tumor cells or tissues into a host animal. Given the complexity and unique characteristics of each tumor, PDXs are models of choice in cancer research and precision medicine. In this context, the zebrafish PDX model (zPDX or zAvatar) has been recognized as a promising in vivo model to directly challenge patient cells with anti-cancer therapies in a personalized manner. The assay relies on the injection of tumor cells from patients into zebrafish embryos to then test and identify the best available drug combination for a particular patient. Compared to mouse PDXs, zAvatar assays take less time and do not require in vitro or in vivo cell expansion. The present article describes how to generate zAvatars from resected digestive cancer from surgeries and how to then use them for anti-cancer therapy screening. We describe the steps for tumor sample collection and cryopreservation, sample preparation and fluorescent labeling for microinjection into zebrafish embryos, drug administration, and analysis of tumor behavior by single-cell confocal imaging. We provide detailed protocols and helpful tips for performing this assay, and we address the technical challenges associated with the workflow. © 2022 Wiley Periodicals LLC. Basic Protocol 1: Patient tumor sample collection and cryopreservation Basic Protocol 2: Generation of zAvatars and anti-cancer treatment Basic Protocol 3: Whole-mount immunofluorescence Basic Protocol 4: Confocal imaging and analysis.
Collapse
Affiliation(s)
- Bruna Costa
- Champalimaud Foundation, Champalimaud Research, Lisbon, Portugal
| | - Marta F Estrada
- Champalimaud Foundation, Champalimaud Research, Lisbon, Portugal
| | | | - Rita Fior
- Champalimaud Foundation, Champalimaud Research, Lisbon, Portugal
| |
Collapse
|
31
|
Hou J, He Z, Liu T, Chen D, Wang B, Wen Q, Zheng X. Evolution of Molecular Targeted Cancer Therapy: Mechanisms of Drug Resistance and Novel Opportunities Identified by CRISPR-Cas9 Screening. Front Oncol 2022; 12:755053. [PMID: 35372044 PMCID: PMC8970599 DOI: 10.3389/fonc.2022.755053] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/17/2022] [Indexed: 12/14/2022] Open
Abstract
Molecular targeted therapy has revolutionized the landscape of cancer treatment due to better therapeutic responses and less systemic toxicity. However, therapeutic resistance is a major challenge in clinical settings that hinders continuous clinical benefits for cancer patients. In this regard, unraveling the mechanisms of drug resistance may identify new druggable genetic alterations for molecularly targeted therapies, thus contributing to improved therapeutic efficacies. The recent rapid development of novel methodologies including CRISPR-Cas9 screening technology and patient-derived models provides powerful tools to dissect the underlying mechanisms of resistance to targeted cancer therapies. In this review, we updated therapeutic targets undergoing preclinical and clinical evaluation for various cancer types. More importantly, we provided comprehensive elaboration of high throughput CRISPR-Cas9 screening in deciphering potential mechanisms of unresponsiveness to molecularly targeted therapies, which will shed light on the discovery of novel opportunities for designing next-generation anti-cancer drugs.
Collapse
Affiliation(s)
- Jue Hou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zongsheng He
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Tian Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Dongfeng Chen
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bin Wang
- Department of Gastroenterology, Chongqing Key Laboratory of Digestive Malignancies, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xi Zheng
- Department of Gastroenterology, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
32
|
Bailone RL, Fukushima HCS, de Aguiar LK, Borra RC. The endocannabinoid system in zebrafish and its potential to study the effects of Cannabis in humans. Lab Anim Res 2022; 38:5. [PMID: 35193700 PMCID: PMC8862295 DOI: 10.1186/s42826-022-00116-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/14/2022] [Indexed: 12/19/2022] Open
Abstract
Zebrafish is considered an unprecedented animal model in drug discovery. A review of the literature presents highlights and elucidates the biological effects of chemical components found in Cannabis sativa. Particular attention is paid to endocannabinoid system (eCB) and its main receptors (CB1 and CB2). The zebrafish model is a promising one for the study of cannabinoids because of the many similarities to the human system. Despite the recent advances on the eCB system, there is still the need to elucidate some of the interactions and, thus, the zebrafish model can be used for that purpose as it respects the 3Rs concept and reduced time and costs. In view of the relevance of cannabinoids in the treatment and prevention of diseases, as well as the importance of the zebrafish animal model in elucidating the biological effects of new drugs, the aim of this study was to bring to light information on the use of the zebrafish animal model in testing C. sativa-based medicines.
Collapse
|
33
|
Chen S, Jiao Y, Pan F, Guan Z, Cheng SH, Sun D. Knock-in of a Large Reporter Gene via the High-Throughput Microinjection of the CRISPR/Cas9 System. IEEE Trans Biomed Eng 2022; 69:2524-2532. [PMID: 35133958 DOI: 10.1109/tbme.2022.3149530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The non-viral delivery of the prokaryotic clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9) nuclease system provides promising solutions for gene therapy. However, traditional chemical and physical delivery approaches for gene knock-in are confronted by significant challenges to overcome the draw-backs of low efficiency and high toxicity. An alternative method for directly delivering CRISPR components into single cells is microinjection. Here, we present the high-throughput robotic microinjection of CRISPR machinery plasmids to produce gene insertions. We demonstrate that the microinjection of CRISPR/Cas9 with an enhanced green fluorescent protein (eGFP) donor template into single HepG2 cells can achieve re-porter gene knock-in targeting the adeno-associated virus site 1 locus. Homology-directed repair-mediated knock-in can be ob-served with an efficiency of 41%. Assessment via T7E1 assay indicates that the eGFP knock-in cells exhibit no detectable changes at potential off-target sites. A case study of injecting the eGFP knock-in cells into zebrafish (Danio rerio) embryos to form an in vivo tumor model is conducted. Results demonstrate the efficiency of combining microinjection with the CRISPR/Cas9 system in achieving gene knock-in.
Collapse
|
34
|
Weiss JM, Lumaquin-Yin D, Montal E, Suresh S, Leonhardt CS, White RM. Shifting the focus of zebrafish toward a model of the tumor microenvironment. eLife 2022; 11:69703. [PMID: 36538362 PMCID: PMC9767465 DOI: 10.7554/elife.69703] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/21/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer cells exist in a complex ecosystem with numerous other cell types in the tumor microenvironment (TME). The composition of this tumor/TME ecosystem will vary at each anatomic site and affects phenotypes such as initiation, metastasis, and drug resistance. A mechanistic understanding of the large number of cell-cell interactions between tumor and TME requires models that allow us to both characterize as well as genetically perturb this complexity. Zebrafish are a model system optimized for this problem, because of the large number of existing cell-type-specific drivers that can label nearly any cell in the TME. These include stromal cells, immune cells, and tissue resident normal cells. These cell-type-specific promoters/enhancers can be used to drive fluorophores to facilitate imaging and also CRISPR cassettes to facilitate perturbations. A major advantage of the zebrafish is the ease by which large numbers of TME cell types can be studied at once, within the same animal. While these features make the zebrafish well suited to investigate the TME, the model has important limitations, which we also discuss. In this review, we describe the existing toolset for studying the TME using zebrafish models of cancer and highlight unique biological insights that can be gained by leveraging this powerful resource.
Collapse
Affiliation(s)
- Joshua M Weiss
- Weill-Cornel Medical College, Tri-Institutional M.D./Ph.D. ProgramNew YorkUnited States
| | - Dianne Lumaquin-Yin
- Weill-Cornel Medical College, Tri-Institutional M.D./Ph.D. ProgramNew YorkUnited States
| | - Emily Montal
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Shruthy Suresh
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Carl S Leonhardt
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Richard M White
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States,Department of Medicine, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| |
Collapse
|
35
|
An Orthotopic Model of Uveal Melanoma in Zebrafish Embryo: A Novel Platform for Drug Evaluation. Biomedicines 2021; 9:biomedicines9121873. [PMID: 34944689 PMCID: PMC8698893 DOI: 10.3390/biomedicines9121873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/30/2021] [Accepted: 12/07/2021] [Indexed: 12/19/2022] Open
Abstract
Uveal melanoma is a highly metastatic tumor, representing the most common primary intraocular malignancy in adults. Tumor cell xenografts in zebrafish embryos may provide the opportunity to study in vivo different aspects of the neoplastic disease and its response to therapy. Here, we established an orthotopic model of uveal melanoma in zebrafish by injecting highly metastatic murine B16-BL6 and B16-LS9 melanoma cells, human A375M melanoma cells, and human 92.1 uveal melanoma cells into the eye of zebrafish embryos in the proximity of the developing choroidal vasculature. Immunohistochemical and immunofluorescence analyses showed that melanoma cells proliferate during the first four days after injection and move towards the eye surface. Moreover, bioluminescence analysis of luciferase-expressing human 92.1 uveal melanoma cells allowed the quantitative assessment of the antitumor activity exerted by the canonical chemotherapeutic drugs paclitaxel, panobinostat, and everolimus after their injection into the grafted eye. Altogether, our data demonstrate that the zebrafish embryo eye is a permissive environment for the growth of invasive cutaneous and uveal melanoma cells. In addition, we have established a new luciferase-based in vivo orthotopic model that allows the quantification of human uveal melanoma cells engrafted in the zebrafish embryo eye, and which may represent a suitable tool for the screening of novel drug candidates for uveal melanoma therapy.
Collapse
|
36
|
Mendonça-Gomes JM, Valverde TM, Martins TMDM, Charlie-Silva I, Padovani BN, Fénero CM, da Silva EM, Domingues RZ, Melo-Hoyos DC, Corrêa-Junior JD, Câmara NOS, Góes AM, Gomes DA. Long-term dexamethasone treatment increases the engraftment efficiency of human breast cancer cells in adult zebrafish. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100007. [DOI: 10.1016/j.fsirep.2021.100007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 01/03/2023] Open
|
37
|
Nathan J, Ramachandran A. Efficacy of marine biomolecules on angiogenesis by targeting hypoxia inducible factor/vascular endothelial growth factor signaling in zebrafish model. J Biochem Mol Toxicol 2021; 36:e22954. [PMID: 34783123 DOI: 10.1002/jbt.22954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/10/2021] [Accepted: 11/01/2021] [Indexed: 12/22/2022]
Abstract
Marine resources are notably explored for their unique biomolecules that have been designed to be drug targets for their immense potential against various pathologies. These biomolecules are mostly secondary metabolites from different species that include sponges, tunicates, echinoderms, ascidians, algae, and marine symbionts. Among the various biological activities of the marine biomolecules, antiangiogenic property has gained much significance in alternate therapy for treatment against cancer. Hypoxia inducible factor (HIF) and vascular endothelial growth factor (VEGF) are the prime signaling pathways related to angiogenesis that are exclusively designated as markers for critical selection of novel inhibitors. This is mainly due to their importance in tumor induction and regulatory control over other interlinked pathways involved in cancer. Small molecular drug screening using the zebrafish model has been an advantage in cancer research in recent times. This review addresses the importance of marine biomolecules and their antiangiogenic efficacy by targeting HIF/VEGF pathways experimented in the zebrafish model in the last decade. Thus, it would provide more clear insights into the role of biomolecules in alternative cancer therapy.
Collapse
Affiliation(s)
- Jhansi Nathan
- Zebrafish Developmental Biology Laboratory, AUKBC Research Centre, Anna University, Chennai, India
| | | |
Collapse
|
38
|
Molina B, Chavez J, Grainger S. Zebrafish models of acute leukemias: Current models and future directions. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2021; 10:e400. [PMID: 33340278 PMCID: PMC8213871 DOI: 10.1002/wdev.400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 12/19/2022]
Abstract
Acute myeloid leukemias (AML) and acute lymphoid leukemias (ALL) are heterogenous diseases encompassing a wide array of genetic mutations with both loss and gain of function phenotypes. Ultimately, these both result in the clonal overgrowth of blast cells in the bone marrow, peripheral blood, and other tissues. As a consequence of this, normal hematopoietic stem cell function is severely hampered. Technologies allowing for the early detection of genetic alterations and understanding of these varied molecular pathologies have helped to advance our treatment regimens toward personalized targeted therapies. In spite of this, both AML and ALL continue to be a major cause of morbidity and mortality worldwide, in part because molecular therapies for the plethora of genetic abnormalities have not been developed. This underscores the current need for better model systems for therapy development. This article reviews the current zebrafish models of AML and ALL and discusses how novel gene editing tools can be implemented to generate better models of acute leukemias. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cells and Disease Technologies > Perturbing Genes and Generating Modified Animals.
Collapse
Affiliation(s)
- Brandon Molina
- Biology Department, San Diego State University, San Diego, California, USA
| | - Jasmine Chavez
- Biology Department, San Diego State University, San Diego, California, USA
| | - Stephanie Grainger
- Biology Department, San Diego State University, San Diego, California, USA
| |
Collapse
|
39
|
Lu X, Yan G, Klauck SM, Fleischer E, Klinger A, Sugimoto Y, Shan L, Efferth T. Cytotoxicity of 4-hydroxy-N-(naphthalen-1-yl)-2-oxo-2H-chromene-3-carboxamide in multidrug-resistant cancer cells through activation of PERK/eIF2α/ATF4 pathway. Biochem Pharmacol 2021; 193:114788. [PMID: 34582772 DOI: 10.1016/j.bcp.2021.114788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 02/06/2023]
Abstract
After decades of research, multidrug resistance (MDR) remains a huge challenge in cancer treatment. In this study, the cytotoxic of 4-hydroxy-N-(naphthalen-1-yl)-2-oxo-2H-chromene-3-carboxamide (MCC1734) has been investigated towards multidrug-resistant cancer cell lines. MCC1734 exerted cytotoxicity on cell lines expressing different mechanisms of drug resistance (P-glycoprotein, BCRP, ABCB5, EGFR, p53 knockout) to a different extent. Interestingly, sensitive CCRF-CEM cells and multidrug-resistant P-gp-overexpressing CEM/ADR5000 cells represented similar sensitivity towards MCC1734, indicating MCC1734 can bypass P-gp-mediated resistance. Microarray-based mRNA expression revealed that MCC1734 affected cells by multiple pathways, including cell cycle regulation, mitochondrial dysfunction, apoptosis signaling, and EIF2 signaling. MCC1734 stimulated the generation of excessive reactive oxygen species and the collapse of mitochondria membrane potential in CCRF-CEM cells, companied by the arrest of the cell cycle in the G2M phase and apoptosis induction as determined by flow cytometry. In addition, our immunoblotting analysis highlighted that MCC1734 triggered endoplasmic reticulum (ER) stress, evidenced by the activation of p-PERK, p-eIF2α, ATF4 and CHOP. The anti-cancer effects of MCC1734 were further observed in vivo using human xenograft tumors transplanted to zebrafish, providing further support for MCC1734 as a promising new candidate for cancer drug development.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Ge Yan
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | | | | | - Yoshikazu Sugimoto
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
40
|
Umans RA, Ten Kate M, Pollock C, Sontheimer H. Fishing for Contact: Modeling Perivascular Glioma Invasion in the Zebrafish Brain. ACS Pharmacol Transl Sci 2021; 4:1295-1305. [PMID: 34423267 DOI: 10.1021/acsptsci.0c00129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly invasive, central nervous system (CNS) cancer for which there is no cure. Invading tumor cells evade treatment, limiting the efficacy of the current standard of care regimen. Understanding the underlying invasive behaviors that support tumor growth may allow for generation of novel GBM therapies. Zebrafish (Danio rerio) are attractive for genetics and live imaging and have, in recent years, emerged as a model system suitable for cancer biology research. While other groups have studied CNS tumors using zebrafish, few have concentrated on the invasive behaviors supporting the development of these diseases. Previous studies demonstrated that one of the main mechanisms of GBM invasion is perivascular invasion, i.e., single tumor cell migration along blood vessels. Here, we characterize phenotypes, methodology, and potential therapeutic avenues for utilizing zebrafish to model perivascular GBM invasion. Using patient-derived xenolines or an adherent cell line, we demonstrate tumor expansion within the zebrafish brain. Within 24-h postintracranial injection, D54-MG-tdTomato glioma cells produce fingerlike projections along the zebrafish brain vasculature. As few as 25 GBM cells were sufficient to promote single cell vessel co-option. Of note, these tumor-vessel interactions are CNS specific and do not occur on pre-existing blood vessels when injected into the animal's peripheral tissue. Tumor-vessel interactions increase over time and can be pharmacologically disrupted through inhibition of Wnt signaling. Therefore, zebrafish serve as a favorable model system to study perivascular glioma invasion, one of the deadly characteristics that make GBM so difficult to treat.
Collapse
Affiliation(s)
- Robyn A Umans
- Center for Glial Biology in Health, Disease, and Cancer, The Fralin Biomedical Research Institute at VTC, Roanoke, Virginia 24016, United States
| | - Mattie Ten Kate
- School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| | - Carolyn Pollock
- School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| | - Harald Sontheimer
- Center for Glial Biology in Health, Disease, and Cancer, The Fralin Biomedical Research Institute at VTC, Roanoke, Virginia 24016, United States.,School of Neuroscience, Virginia Tech, Sandy Hall, 210 Drillfield Drive, Blacksburg, Virginia 24061, United States
| |
Collapse
|
41
|
Letrado P, Mole H, Montoya M, Palacios I, Barriuso J, Hurlstone A, Díez-Martínez R, Oyarzabal J. Systematic Roadmap for Cancer Drug Screening Using Zebrafish Embryo Xenograft Cancer Models: Melanoma Cell Line as a Case Study. Cancers (Basel) 2021; 13:cancers13153705. [PMID: 34359605 PMCID: PMC8345186 DOI: 10.3390/cancers13153705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Currently, there is no consensus in the scientific literature regarding the zebrafish embryo xenotransplantation procedure for drug screening. Thus, this study sets systematic guidelines for maximizing the reproducibility of drug screening in zebrafish-embryo cancer xenograft models based on evaluating every step of the procedure in a real case scenario in which the chemical properties of the compounds are unknown or not optimal. It aims to be a stepping stone to bring the versatility of zebrafish embryos to drug screening for cancer. The present work helps our group to pursue the objective of establishing zebrafish embryos as a valuable alternative to mice models; and hopefully, will help other groups in this field to progress in the same direction. Abstract Zebrafish embryo tumor transplant models are widely utilized in cancer research. Compared with traditional murine models, the small size and transparency of zebrafish embryos combined with large clutch sizes that increase statistical power and cheap husbandry make them a cost-effective and versatile tool for in vivo drug discovery. However, the lack of a comprehensive analysis of key factors impacting the successful use of these models impedes the establishment of basic guidelines for systematic screening campaigns. Thus, we explored the following crucial factors: (i) user-independent inclusion criteria, focusing on sample homogeneity; (ii) metric definition for data analysis; (iii) tumor engraftment criteria; (iv) image analysis versus quantification of human cancer cells using qPCR (RNA and gDNA); (v) tumor implantation sites; (vi) compound distribution (intratumoral administration versus alternative inoculation sites); and (vii) efficacy (intratumoral microinjection versus compound solution in media). Based on these analyses and corresponding assessments, we propose the first roadmap for systematic drug discovery screening in zebrafish xenograft cancer models using a melanoma cell line as a case study. This study aims to help the wider cancer research community to consider the adoption of this versatile model for cancer drug screening projects.
Collapse
Affiliation(s)
- Patricia Letrado
- Ikan Biotech SL, Centro Europeo de Empresas e Innovación de Navarra (CEIN), 31110 Noain, Spain;
- Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Correspondence: (P.L.); (J.O.)
| | - Holly Mole
- Division of Cancer Sciences, School of Medical Sciences, The University of Manchester, Manchester M13 9PL, UK; (H.M.); (J.B.)
| | - María Montoya
- Cellomics Unit, Spanish National Center for Cardiovascular Research (CNIC), 28029 Madrid, Spain; (M.M.); (I.P.)
| | - Irene Palacios
- Cellomics Unit, Spanish National Center for Cardiovascular Research (CNIC), 28029 Madrid, Spain; (M.M.); (I.P.)
| | - Jorge Barriuso
- Division of Cancer Sciences, School of Medical Sciences, The University of Manchester, Manchester M13 9PL, UK; (H.M.); (J.B.)
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Adam Hurlstone
- Division of Infection, Immunology and Respiratory Medicine, School of Biological Science, The University of Manchester, Manchester M13 9PT, UK;
| | - Roberto Díez-Martínez
- Ikan Biotech SL, Centro Europeo de Empresas e Innovación de Navarra (CEIN), 31110 Noain, Spain;
| | - Julen Oyarzabal
- Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Correspondence: (P.L.); (J.O.)
| |
Collapse
|
42
|
Bozzer S, Bo MD, Toffoli G, Macor P, Capolla S. Nanoparticles-Based Oligonucleotides Delivery in Cancer: Role of Zebrafish as Animal Model. Pharmaceutics 2021; 13:1106. [PMID: 34452067 PMCID: PMC8400075 DOI: 10.3390/pharmaceutics13081106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/29/2022] Open
Abstract
Oligonucleotide (ON) therapeutics are molecular target agents composed of chemically synthesized DNA or RNA molecules capable of inhibiting gene expression or protein function. How ON therapeutics can efficiently reach the inside of target cells remains a problem still to be solved in the majority of potential clinical applications. The chemical structure of ON compounds could affect their capability to pass through the plasma membrane. Other key factors are nuclease degradation in the extracellular space, renal clearance, reticulo-endothelial system, and at the target cell level, the endolysosomal system and the possible export via exocytosis. Several delivery platforms have been proposed to overcome these limits including the use of lipidic, polymeric, and inorganic nanoparticles, or hybrids between them. The possibility of evaluating the efficacy of the proposed therapeutic strategies in useful in vivo models is still a pivotal need, and the employment of zebrafish (ZF) models could expand the range of possibilities. In this review, we briefly describe the main ON therapeutics proposed for anticancer treatment, and the different strategies employed for their delivery to cancer cells. The principal features of ZF models and the pros and cons of their employment in the development of ON-based therapeutic strategies are also discussed.
Collapse
Affiliation(s)
- Sara Bozzer
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (M.D.B.); (G.T.); (S.C.)
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (M.D.B.); (G.T.); (S.C.)
| | - Paolo Macor
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Sara Capolla
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; (M.D.B.); (G.T.); (S.C.)
| |
Collapse
|
43
|
Lu X, Yan G, Dawood M, Klauck SM, Sugimoto Y, Klinger A, Fleischer E, Shan L, Efferth T. A novel moniliformin derivative as pan-inhibitor of histone deacetylases triggering apoptosis of leukemia cells. Biochem Pharmacol 2021; 194:114677. [PMID: 34265280 DOI: 10.1016/j.bcp.2021.114677] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022]
Abstract
New and potent agents that evade multidrug resistance (MDR) and inhibit epigenetic modifications are of great interest in cancer drug development. Here, we describe that a moniliformin derivative (IUPAC name: 3-(naphthalen-2-ylsulfanyl)-4-{[(2Z)-1,3,3-trimethyl-2,3-dihydro-1H-indol-2-ylidene]methyl}cyclobut-3-ene-1,2-dione; code: MCC1381) bypasses P-gp-mediated MDR. Using transcriptomics, we identified a large number of genes significantly regulated in response to MCC1381, which affected the cell cycle and disturbed cellular death and survival. The potential targets of MCC1381 might be histone deacetylases (HDACs) as predicted by SwissTargetPrediction. In silico studies confirmed that MCC1381 presented comparable affinity with HDAC1, 2, 3, 6, 8 and 11. Besides, the inhibition activity of HDACs was dose-dependently inhibited by MCC1381. Particularly, a strong binding affinity was observed between MCC1381 and HDAC6 by microscale thermophoresis analysis. MCC1381 decreased the expression of HDAC6, inversely correlated with the increase of acetylated HDAC6 substrates, acetylation p53 and α-tubulin. Furthermore, MCC1381 arrested the cell cycle at the G2/M phase, induced the generation of reactive oxygen species and collapse of the mitochondrial membrane potential. MCC1381 exhibited in vivo anti-cancer activity in xenografted zebrafish. Collectively, MCC1381 extended cytotoxicity towards P-gp-resistant leukemia cancer cells and may act as a pan-HDACs inhibitor, indicating that MCC1381 is a novel candidate for cancer therapy.
Collapse
Affiliation(s)
- Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Ge Yan
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Mona Dawood
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany; Department of Molecular Biology, Faculty of Medical Laboratory Sciences, Al-Neelain University, Khartoum, Sudan
| | - Sabine M Klauck
- Division of Cancer Genome Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Yoshikazu Sugimoto
- Division of Chemotherapy, Faculty of Pharmacy, Keio University, Tokyo, Japan
| | | | | | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
44
|
Munekata PES, Pateiro M, Conte-Junior CA, Domínguez R, Nawaz A, Walayat N, Movilla Fierro E, Lorenzo JM. Marine Alkaloids: Compounds with In Vivo Activity and Chemical Synthesis. Mar Drugs 2021; 19:374. [PMID: 34203532 PMCID: PMC8306672 DOI: 10.3390/md19070374] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/24/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022] Open
Abstract
Marine alkaloids comprise a class of compounds with several nitrogenated structures that can be explored as potential natural bioactive compounds. The scientific interest in these compounds has been increasing in the last decades, and many studies have been published elucidating their chemical structure and biological effects in vitro. Following this trend, the number of in vivo studies reporting the health-related properties of marine alkaloids has been increasing and providing more information about the effects in complex organisms. Experiments with animals, especially mice and zebrafish, are revealing the potential health benefits against cancer development, cardiovascular diseases, seizures, Alzheimer's disease, mental health disorders, inflammatory diseases, osteoporosis, cystic fibrosis, oxidative stress, human parasites, and microbial infections in vivo. Although major efforts are still necessary to increase the knowledge, especially about the translation value of the information obtained from in vivo experiments to clinical trials, marine alkaloids are promising candidates for further experiments in drug development.
Collapse
Affiliation(s)
- Paulo E. S. Munekata
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Mirian Pateiro
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Carlos A. Conte-Junior
- Centro de Tecnologia, Programa de Pós-Graduação em Ciência de Alimentos, Instituto de Química, Universidade Federal do Rio de Janeiro, Avenida Athos da Silveira Ramos 149, Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil;
| | - Rubén Domínguez
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
| | - Asad Nawaz
- Jiangsu Key Laboratory of Crop Genetics and Physiology, College of Agriculture, Yangzhou University, Yangzhou 225009, China;
| | - Noman Walayat
- Department of Food Science and Engineering, College of Ocean, Zhejiang University of Technology, Hangzhou 310014, China;
| | | | - José M. Lorenzo
- Centro Tecnológico de la Carne de Galicia, Parque Tecnológico de Galicia, rúa Galicia No. 4, San Cibrao das Viñas, 32900 Ourense, Spain; (P.E.S.M.); (M.P.); (R.D.)
- Área de Tecnología de los Alimentos, Facultad de Ciencias de Ourense, Universidad de Vigo, 32004 Ourense, Spain
| |
Collapse
|
45
|
Sajjad H, Imtiaz S, Noor T, Siddiqui YH, Sajjad A, Zia M. Cancer models in preclinical research: A chronicle review of advancement in effective cancer research. Animal Model Exp Med 2021; 4:87-103. [PMID: 34179717 PMCID: PMC8212826 DOI: 10.1002/ame2.12165] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/04/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer is a major stress for public well-being and is the most dreadful disease. The models used in the discovery of cancer treatment are continuously changing and extending toward advanced preclinical studies. Cancer models are either naturally existing or artificially prepared experimental systems that show similar features with human tumors though the heterogeneous nature of the tumor is very familiar. The choice of the most fitting model to best reflect the given tumor system is one of the real difficulties for cancer examination. Therefore, vast studies have been conducted on the cancer models for developing a better understanding of cancer invasion, progression, and early detection. These models give an insight into cancer etiology, molecular basis, host tumor interaction, the role of microenvironment, and tumor heterogeneity in tumor metastasis. These models are also used to predict novel cancer markers, targeted therapies, and are extremely helpful in drug development. In this review, the potential of cancer models to be used as a platform for drug screening and therapeutic discoveries are highlighted. Although none of the cancer models is regarded as ideal because each is associated with essential caveats that restraint its application yet by bridging the gap between preliminary cancer research and translational medicine. However, they promise a brighter future for cancer treatment.
Collapse
Affiliation(s)
- Humna Sajjad
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Saiqa Imtiaz
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Tayyaba Noor
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| | | | - Anila Sajjad
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| | - Muhammad Zia
- Department of BiotechnologyQuaid‐i‐Azam UniversityIslamabadPakistan
| |
Collapse
|
46
|
Zampedri C, Martínez-Flores WA, Melendez-Zajgla J. The Use of Zebrafish Xenotransplant Assays to Analyze the Role of lncRNAs in Breast Cancer. Front Oncol 2021; 11:687594. [PMID: 34123857 PMCID: PMC8190406 DOI: 10.3389/fonc.2021.687594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/04/2021] [Indexed: 12/19/2022] Open
Abstract
Breast cancer represents a great challenge since it is the first cause of death by cancer in women worldwide. LncRNAs are a newly described class of non-coding RNAs that participate in cancer progression. Their use as cancer markers and possible therapeutic targets has recently gained strength. Animal xenotransplants allows for in vivo monitoring of disease development, molecular elucidation of pathogenesis and the design of new therapeutic strategies. Nevertheless, the cost and complexities of mice husbandry makes medium to high throughput assays difficult. Zebrafishes (Danio rerio) represent a novel model for these assays, given the ease with which xenotransplantation trials can be performed and the economic and experimental advantages it offers. In this review we propose the use of xenotransplants in zebrafish to study the role of breast cancer lncRNAs using low to medium high throughput assays.
Collapse
Affiliation(s)
- Cecilia Zampedri
- Functional Genomics Laboratories, Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| | | | - Jorge Melendez-Zajgla
- Functional Genomics Laboratories, Instituto Nacional de Medicina Genomica, Mexico City, Mexico
| |
Collapse
|
47
|
Zebrafish, an In Vivo Platform to Screen Drugs and Proteins for Biomedical Use. Pharmaceuticals (Basel) 2021; 14:ph14060500. [PMID: 34073947 PMCID: PMC8225009 DOI: 10.3390/ph14060500] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/14/2021] [Accepted: 05/20/2021] [Indexed: 12/28/2022] Open
Abstract
The nearly simultaneous convergence of human genetics and advanced molecular technologies has led to an improved understanding of human diseases. At the same time, the demand for drug screening and gene function identification has also increased, albeit time- and labor-intensive. However, bridging the gap between in vitro evidence from cell lines and in vivo evidence, the lower vertebrate zebrafish possesses many advantages over higher vertebrates, such as low maintenance, high fecundity, light-induced spawning, transparent embryos, short generation interval, rapid embryonic development, fully sequenced genome, and some phenotypes similar to human diseases. Such merits have popularized the zebrafish as a model system for biomedical and pharmaceutical studies, including drug screening. Here, we reviewed the various ways in which zebrafish serve as an in vivo platform to perform drug and protein screening in the fields of rare human diseases, social behavior and cancer studies. Since zebrafish mutations faithfully phenocopy many human disorders, many compounds identified from zebrafish screening systems have advanced to early clinical trials, such as those for Adenoid cystic carcinoma, Dravet syndrome and Diamond-Blackfan anemia. We also reviewed and described how zebrafish are used to carry out environmental pollutant detection and assessment of nanoparticle biosafety and QT prolongation.
Collapse
|
48
|
Miao KZ, Kim GY, Meara GK, Qin X, Feng H. Tipping the Scales With Zebrafish to Understand Adaptive Tumor Immunity. Front Cell Dev Biol 2021; 9:660969. [PMID: 34095125 PMCID: PMC8173129 DOI: 10.3389/fcell.2021.660969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/19/2021] [Indexed: 12/20/2022] Open
Abstract
The future of improved immunotherapy against cancer depends on an in-depth understanding of the dynamic interactions between the immune system and tumors. Over the past two decades, the zebrafish has served as a valuable model system to provide fresh insights into both the development of the immune system and the etiologies of many different cancers. This well-established foundation of knowledge combined with the imaging and genetic capacities of the zebrafish provides a new frontier in cancer immunology research. In this review, we provide an overview of the development of the zebrafish immune system along with a side-by-side comparison of its human counterpart. We then introduce components of the adaptive immune system with a focus on their roles in the tumor microenvironment (TME) of teleosts. In addition, we summarize zebrafish models developed for the study of cancer and adaptive immunity along with other available tools and technology afforded by this experimental system. Finally, we discuss some recent research conducted using the zebrafish to investigate adaptive immune cell-tumor interactions. Without a doubt, the zebrafish will arise as one of the driving forces to help expand the knowledge of tumor immunity and facilitate the development of improved anti-cancer immunotherapy in the foreseeable future.
Collapse
Affiliation(s)
- Kelly Z Miao
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Grace Y Kim
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Grace K Meara
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Xiaodan Qin
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States
| | - Hui Feng
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, Boston, MA, United States.,Department of Medicine, Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
49
|
Saraiva SM, Gutiérrez-Lovera C, Martínez-Val J, Lores S, Bouzo BL, Díez-Villares S, Alijas S, Pensado-López A, Vázquez-Ríos AJ, Sánchez L, de la Fuente M. Edelfosine nanoemulsions inhibit tumor growth of triple negative breast cancer in zebrafish xenograft model. Sci Rep 2021; 11:9873. [PMID: 33972572 PMCID: PMC8110995 DOI: 10.1038/s41598-021-87968-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/29/2021] [Indexed: 12/18/2022] Open
Abstract
Triple negative breast cancer (TNBC) is known for being very aggressive, heterogeneous and highly metastatic. The standard of care treatment is still chemotherapy, with adjacent toxicity and low efficacy, highlighting the need for alternative and more effective therapeutic strategies. Edelfosine, an alkyl-lysophospholipid, has proved to be a promising therapy for several cancer types, upon delivery in lipid nanoparticles. Therefore, the objective of this work was to explore the potential of edelfosine for the treatment of TNBC. Edelfosine nanoemulsions (ET-NEs) composed by edelfosine, Miglyol 812 and phosphatidylcholine as excipients, due to their good safety profile, presented an average size of about 120 nm and a neutral zeta potential, and were stable in biorelevant media. The ability of ET-NEs to interrupt tumor growth in TNBC was demonstrated both in vitro, using a highly aggressive and invasive TNBC cell line, and in vivo, using zebrafish embryos. Importantly, ET-NEs were able to penetrate through the skin barrier of MDA-MB 231 xenografted zebrafish embryos, into the yolk sac, leading to an effective decrease of highly aggressive and invasive tumoral cells' proliferation. Altogether the results demonstrate the potential of ET-NEs for the development of new therapeutic approaches for TNBC.
Collapse
Affiliation(s)
- Sofia M Saraiva
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Carlha Gutiérrez-Lovera
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - Jeannette Martínez-Val
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - Sainza Lores
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Belén L Bouzo
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Sandra Díez-Villares
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Sandra Alijas
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
| | - Alba Pensado-López
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Abi Judit Vázquez-Ríos
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain
- Cancer Network Research (CIBERONC), Madrid, Spain
| | - Laura Sánchez
- Department of Zoology, Genetics and Physical Anthropology, Campus of Lugo, University of Santiago de Compostela, Lugo, Spain
| | - María de la Fuente
- Nano-Oncology and Translational Therapeutics Unit, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), SERGAS, Santiago de Compostela, Spain.
- Cancer Network Research (CIBERONC), Madrid, Spain.
| |
Collapse
|
50
|
Monroe JD, Basheer F, Gibert Y. Xmrks the Spot: Fish Models for Investigating Epidermal Growth Factor Receptor Signaling in Cancer Research. Cells 2021; 10:1132. [PMID: 34067095 PMCID: PMC8150686 DOI: 10.3390/cells10051132] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 12/29/2022] Open
Abstract
Studies conducted in several fish species, e.g., Xiphophorus hellerii (green swordtail) and Xiphophorus maculatus (southern platyfish) crosses, Oryzias latipes (medaka), and Danio rerio (zebrafish), have identified an oncogenic role for the receptor tyrosine kinase, Xmrk, a gene product closely related to the human epidermal growth factor receptor (EGFR), which is associated with a wide variety of pathological conditions, including cancer. Comparative analyses of Xmrk and EGFR signal transduction in melanoma have shown that both utilize STAT5 signaling to regulate apoptosis and cell proliferation, PI3K to modulate apoptosis, FAK to control migration, and the Ras/Raf/MEK/MAPK pathway to regulate cell survival, proliferation, and differentiation. Further, Xmrk and EGFR may also modulate similar chemokine, extracellular matrix, oxidative stress, and microRNA signaling pathways in melanoma. In hepatocellular carcinoma (HCC), Xmrk and EGFR signaling utilize STAT5 to regulate cell proliferation, and Xmrk may signal through PI3K and FasR to modulate apoptosis. At the same time, both activate the Ras/Raf/MEK/MAPK pathway to regulate cell proliferation and E-cadherin signaling. Xmrk models of melanoma have shown that inhibitors of PI3K and MEK have an anti-cancer effect, and in HCC, that the steroidal drug, adrenosterone, can prevent metastasis and recover E-cadherin expression, suggesting that fish Xmrk models can exploit similarities with EGFR signal transduction to identify and study new chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jerry D. Monroe
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA;
| | - Faiza Basheer
- School of Medicine, Deakin University, Locked Bag 20000, Geelong, VIC 3220, Australia;
| | - Yann Gibert
- Department of Cell and Molecular Biology, Cancer Center and Research Institute, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA;
| |
Collapse
|