1
|
Ventura Fernandes BH, Junqueira MS, MacRae C, Silveira de Carvalho LR. Standardizing CRISPR-Cas13 knockdown technique to investigate the role of cdh2 gene in pituitary development through growth hormone expression and transcription factors. Front Endocrinol (Lausanne) 2024; 15:1466638. [PMID: 39449741 PMCID: PMC11499105 DOI: 10.3389/fendo.2024.1466638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Congenital hypopituitarism (CH) is characterized by the deficiency of pituitary hormones. Among CH patients, 85% lack a molecular diagnosis. Whole Exome Sequencing (WES) identified a homozygous variant (c.865G>A, p.Val289Ile) in the CDH2 gene, responsible for N-Cadherin production, crucial for cell-cell adhesion. Predicted to be likely pathogenic, the variant was found in a patient deficient in GH, TSH, ACTH, and LH/FSH. Its impact on cell adhesion was confirmed in L1 fibroblast cell lines. Objective Create a cdh2 knockdown in zebrafish for investigating its role in pituitary development through growth hormone and transcription factors expression. Methods Utilized pET28B-RfxCas13d-His plasmid for Cas13 mRNA production via in vitro transcription, guiding Cas13 to cdh2 with three RNAs. Injected the complex into single-cell embryos for analysis up to 96 hpf. Assessed gene expression of cdh2, prop1, pit1, and gh1 using RT-qPCR. Evaluated cdh2 protein expression through the western blot technique. Results Knockdown animals displayed developmental delay. The cdh2 expression decreased by 75% within 24 hours, rebounded by 48 hours, and reached wild-type levels by 96 hpf. gh1 expression decreased at 48h but increased by 96 hpf, aligning with WT. No significant differences in prop1 and pit1 expression were observed. Conclusion Our findings underscore cdh2's role in pituitary development and hormonal regulation, offering insights for developmental biology research.
Collapse
Affiliation(s)
- Bianca Helena Ventura Fernandes
- Laboratory of Hormones and Molecular Genetics (LIM/42), Developmental Endocrinology Unit, Discipline of Endocrinology and Metabolism, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Zebrafish Facility, Technical Support Directorate for Teaching and Research, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Mara S. Junqueira
- Center for Translational Research in Oncology, Cancer Institute of the State of São Paulo, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Calum MacRae
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Luciani R. Silveira de Carvalho
- Laboratory of Hormones and Molecular Genetics (LIM/42), Developmental Endocrinology Unit, Discipline of Endocrinology and Metabolism, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Zebrafish Facility, Technical Support Directorate for Teaching and Research, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Sofyantoro F, Septriani NI, Yudha DS, Wicaksono EA, Priyono DS, Putri WA, Primahesa A, Raharjeng ARP, Purwestri YA, Nuringtyas TR. Zebrafish as Versatile Model for Assessing Animal Venoms and Toxins: Current Applications and Future Prospects. Zebrafish 2024; 21:231-242. [PMID: 38608228 DOI: 10.1089/zeb.2023.0088] [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: 04/14/2024] Open
Abstract
Animal venoms and toxins hold promise as sources of novel drug candidates, therapeutic agents, and biomolecules. To fully harness their potential, it is crucial to develop reliable testing methods that provide a comprehensive understanding of their effects and mechanisms of action. However, traditional rodent assays encounter difficulties in mimicking venom-induced effects in human due to the impractical venom dosage levels. The search for reliable testing methods has led to the emergence of zebrafish (Danio rerio) as a versatile model organism for evaluating animal venoms and toxins. Zebrafish possess genetic similarities to humans, rapid development, transparency, and amenability to high-throughput assays, making it ideal for assessing the effects of animal venoms and toxins. This review highlights unique attributes of zebrafish and explores their applications in studying venom- and toxin-induced effects from various species, including snakes, jellyfish, cuttlefish, anemones, spiders, and cone snails. Through zebrafish-based research, intricate physiological responses, developmental alterations, and potential therapeutic interventions induced by venoms are revealed. Novel techniques such as CRISPR/Cas9 gene editing, optogenetics, and high-throughput screening hold great promise for advancing venom research. As zebrafish-based insights converge with findings from other models, the comprehensive understanding of venom-induced effects continues to expand, guiding the development of targeted interventions and promoting both scientific knowledge and practical applications.
Collapse
Affiliation(s)
- Fajar Sofyantoro
- Faculties of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | | | | - Ega Adhi Wicaksono
- Faculties of Agriculture, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Dwi Sendi Priyono
- Faculties of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | | | - Alfian Primahesa
- Faculties of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Anita Restu Puji Raharjeng
- Faculties of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Faculty of Science and Technology, Universitas Islam Negeri Raden Fatah Palembang, South Sumatera, Indonesia
| | - Yekti Asih Purwestri
- Faculties of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Research Center for Biotechnology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Tri Rini Nuringtyas
- Faculties of Biology, Universitas Gadjah Mada, Yogyakarta, Indonesia
- Research Center for Biotechnology, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
3
|
Zizioli D, Quiros-Roldan E, Ferretti S, Mignani L, Tiecco G, Monti E, Castelli F, Zanella I. Dolutegravir and Folic Acid Interaction during Neural System Development in Zebrafish Embryos. Int J Mol Sci 2024; 25:4640. [PMID: 38731859 PMCID: PMC11083492 DOI: 10.3390/ijms25094640] [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: 02/20/2024] [Revised: 04/04/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Dolutegravir (DTG) is one of the most prescribed antiretroviral drugs for treating people with HIV infection, including women of child-bearing potential or pregnant. Nonetheless, neuropsychiatric symptoms are frequently reported. Early reports suggested that, probably in relation to folic acid (FA) shortage, DTG may induce neural tube defects in infants born to women taking the drug during pregnancy. Subsequent reports did not definitively confirm these findings. Recent studies in animal models have highlighted the association between DTG exposure in utero and congenital anomalies, and an increased risk of neurologic abnormalities in children exposed during in utero life has been reported. Underlying mechanisms for DTG-related neurologic symptoms and congenital anomalies are not fully understood. We aimed to deepen our knowledge on the neurodevelopmental effects of DTG exposure and further explore the protective role of FA by the use of zebrafish embryos. We treated embryos at 4 and up to 144 h post fertilization (hpf) with a subtherapeutic DTG concentration (1 μM) and observed the disruption of the anterior-posterior axis and several morphological malformations in the developing brain that were both prevented by pre-exposure (2 hpf) and rescued by post-exposure (10 hpf) with FA. By whole-mount in situ hybridization with riboprobes for genes that are crucial during the early phases of neurodevelopment (ntl, pax2a, ngn1, neurod1) and by in vivo visualization of the transgenic Tg(ngn1:EGFP) zebrafish line, we found that DTG induced severe neurodevelopmental defects over time in most regions of the nervous system (notochord, midbrain-hindbrain boundary, eye, forebrain, midbrain, hindbrain, spinal cord) that were mostly but not completely rescued by FA supplementation. Of note, we observed the disruption of ngn1 expression in the dopaminergic regions of the developing forebrain, spinal cord neurons and spinal motor neuron projections, with the depletion of the tyrosine hydroxylase (TH)+ dopaminergic neurons of the dorsal diencephalon and the strong reduction in larvae locomotion. Our study further supports previous evidence that DTG can interfere with FA pathways in the developing brain but also provides new insights regarding the mechanisms involved in the increased risk of DTG-associated fetal neurodevelopmental defects and adverse neurologic outcomes in in utero exposed children, suggesting the impairment of dopaminergic pathways.
Collapse
Affiliation(s)
- Daniela Zizioli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (D.Z.); (S.F.); (L.M.); (E.M.); (I.Z.)
| | - Eugenia Quiros-Roldan
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (G.T.); (F.C.)
| | - Sara Ferretti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (D.Z.); (S.F.); (L.M.); (E.M.); (I.Z.)
| | - Luca Mignani
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (D.Z.); (S.F.); (L.M.); (E.M.); (I.Z.)
| | - Giorgio Tiecco
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (G.T.); (F.C.)
| | - Eugenio Monti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (D.Z.); (S.F.); (L.M.); (E.M.); (I.Z.)
| | - Francesco Castelli
- Unit of Infectious and Tropical Diseases, Department of Clinical and Experimental Sciences, University of Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy; (G.T.); (F.C.)
| | - Isabella Zanella
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (D.Z.); (S.F.); (L.M.); (E.M.); (I.Z.)
- Cytogenetics and Molecular Genetics Laboratory, Diagnostic Department, ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| |
Collapse
|
4
|
Verma SK, Nandi A, Sinha A, Patel P, Mohanty S, Jha E, Jena S, Kumari P, Ghosh A, Jerman I, Chouhan RS, Dutt A, Samal SK, Mishra YK, Varma RS, Panda PK, Kaushik NK, Singh D, Suar M. The posterity of Zebrafish in paradigm of in vivo molecular toxicological profiling. Biomed Pharmacother 2024; 171:116160. [PMID: 38237351 DOI: 10.1016/j.biopha.2024.116160] [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: 10/04/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
The aggrandised advancement in utility of advanced day-to-day materials and nanomaterials has raised serious concern on their biocompatibility with human and other biotic members. In last few decades, understanding of toxicity of these materials has been given the centre stage of research using many in vitro and in vivo models. Zebrafish (Danio rerio), a freshwater fish and a member of the minnow family has garnered much attention due to its distinct features, which make it an important and frequently used animal model in various fields of embryology and toxicological studies. Given that fertilization and development of zebrafish eggs take place externally, they serve as an excellent model organism for studying early developmental stages. Moreover, zebrafish possess a comparable genetic composition to humans and share almost 70% of their genes with mammals. This particular model organism has become increasingly popular, especially for developmental research. Moreover, it serves as a link between in vitro studies and in vivo analysis in mammals. It is an appealing choice for vertebrate research, when employing high-throughput methods, due to their small size, swift development, and relatively affordable laboratory setup. This small vertebrate has enhanced comprehension of pathobiology and drug toxicity. This review emphasizes on the recent developments in toxicity screening and assays, and the new insights gained about the toxicity of drugs through these assays. Specifically, the cardio, neural, and, hepatic toxicology studies inferred by applications of nanoparticles have been highlighted.
Collapse
Affiliation(s)
- Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar, India.
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Adrija Sinha
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Paritosh Patel
- School of Biotechnology, KIIT University, Bhubaneswar, India; Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897, Seoul, South Korea
| | | | - Ealisha Jha
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Snehasmita Jena
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Puja Kumari
- RECETOX, Faculty of Science, Masaryk University, Kotlarska 2, Brno 61137, Czech Republic
| | - Aishee Ghosh
- School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Ivan Jerman
- National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Raghuraj Singh Chouhan
- Department of Environmental Sciences, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Ateet Dutt
- Instituto de Investigaciones en Materiales, UNAM, CDMX, Mexico
| | - Shailesh Kumar Samal
- Unit of Immunology and Chronic Disease, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, Alsion 2, Sønderborg DK-6400, Denmark
| | - Rajender S Varma
- Institute for Nanomaterials, Advanced Technologies and Innovation (CxI), Technical University of Liberec (TUL), Studentská 1402/2, Liberec 1 461 17, Czech Republic
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897, Seoul, South Korea.
| | - Deobrat Singh
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar, India.
| |
Collapse
|
5
|
Choi SW, Abitbol JM, Cheng AG. Hair Cell Regeneration: From Animals to Humans. Clin Exp Otorhinolaryngol 2024; 17:1-14. [PMID: 38271988 PMCID: PMC10933805 DOI: 10.21053/ceo.2023.01382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/07/2024] [Accepted: 01/17/2024] [Indexed: 01/27/2024] Open
Abstract
Cochlear hair cells convert sound into electrical signals that are relayed via the spiral ganglion neurons to the central auditory pathway. Hair cells are vulnerable to damage caused by excessive noise, aging, and ototoxic agents. Non-mammals can regenerate lost hair cells by mitotic regeneration and direct transdifferentiation of surrounding supporting cells. However, in mature mammals, damaged hair cells are not replaced, resulting in permanent hearing loss. Recent studies have uncovered mechanisms by which sensory organs in non-mammals and the neonatal mammalian cochlea regenerate hair cells, and outlined possible mechanisms why this ability declines rapidly with age in mammals. Here, we review similarities and differences between avian, zebrafish, and mammalian hair cell regeneration. Moreover, we discuss advances and limitations of hair cell regeneration in the mature cochlea and their potential applications to human hearing loss.
Collapse
Affiliation(s)
- Sung-Won Choi
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Otorhinolaryngology-Head and Neck Surgery and Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Pusan National University School of Medicine, Busan, Korea
| | - Julia M. Abitbol
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan G. Cheng
- Department of Otolaryngology-Head and Neck Surgery, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
6
|
Sun J, Wu Q, Wei Y, Zhao W, Lv H, Peng W, Zheng J, Chen Y, Wang Z, Pan Y, Xue Y. Agaricus bisporus-Derived Glucosamine Hydrochloride Regulates VEGF through BMP Signaling to Promote Zebrafish Vascular Development and Impairment Repair. Life (Basel) 2023; 13:2330. [PMID: 38137931 PMCID: PMC10745105 DOI: 10.3390/life13122330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/01/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Glucosamine hydrochloride (GAH) is a natural component of glycoproteins present in almost all human tissues and participates in the construction of human tissues and cell membranes. GAH has a wide range of biological activities, particularly in anti-inflammatory and osteogenic damage repair. At present, little is known about how GAH functions in angiogenesis. To determine the role of GAH on vascular development and impairment repair, we used the inhibitors VRI, DMH1, and dorsomorphin (DM) to construct vascular-impaired models in Tg(kdrl: mCherry) transgenic zebrafish. We then treated with GAH and measured its repair effects on vascular impairment through fluorescence intensity, mRNA, and protein expression levels of vascular-specific markers. Our results indicate that GAH promotes vascular development and repairs impairment by regulating the vascular endothelial growth factor (VEGF) signaling pathway through modulation of bone morphogenetic protein (BMP) signaling. This study provides an experimental basis for the development of GAH as a drug to repair vascular diseases.
Collapse
Affiliation(s)
- Jiarui Sun
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Qici Wu
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Yuxin Wei
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Wei Zhao
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Haokun Lv
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Wei Peng
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Jiayi Zheng
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Yixuan Chen
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Zhengsen Wang
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Yutian Pan
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| | - Yu Xue
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou 363000, China; (J.S.); (Q.W.); (Y.W.); (W.Z.); (H.L.); (W.P.); (J.Z.); (Y.C.); (Z.W.)
- Fujian Fungal Active Substance Engineering Technology Center, Zhangzhou 363000, China
| |
Collapse
|
7
|
Costa FV, Zabegalov KN, Kolesnikova TO, de Abreu MS, Kotova MM, Petersen EV, Kalueff AV. Experimental models of human cortical malformations: from mammals to 'acortical' zebrafish. Neurosci Biobehav Rev 2023; 155:105429. [PMID: 37863278 DOI: 10.1016/j.neubiorev.2023.105429] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
Human neocortex controls and integrates cognition, emotions, perception and complex behaviors. Aberrant cortical development can be triggered by multiple genetic and environmental factors, causing cortical malformations. Animal models, especially rodents, are a valuable tool to probe molecular and physiological mechanisms of cortical malformations. Complementing rodent studies, the zebrafish (Danio rerio) is an important model organism in biomedicine. Although the zebrafish (like other fishes) lacks neocortex, here we argue that this species can still be used to model various aspects and brain phenomena related to human cortical malformations. We also discuss novel perspectives in this field, covering both advantages and limitations of using mammalian and zebrafish models in cortical malformation research. Summarizing mounting evidence, we also highlight the importance of translationally-relevant insights into the pathogenesis of cortical malformations from animal models, and discuss future strategies of research in the field.
Collapse
Affiliation(s)
- Fabiano V Costa
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Konstantin N Zabegalov
- Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | - Tatiana O Kolesnikova
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | | | - Maria M Kotova
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia
| | | | - Allan V Kalueff
- World-class Research Center "Center for Personalized Medicine", Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; Ural Federal University, Yekaterinburg, Russia; Neurobiology Program, Sirius University of Science and Technology, Sirius Federal Territory, Russia.
| |
Collapse
|
8
|
Wynsberghe JV, Vanakker OM. Significance of Premature Vertebral Mineralization in Zebrafish Models in Mechanistic and Pharmaceutical Research on Hereditary Multisystem Diseases. Biomolecules 2023; 13:1621. [PMID: 38002303 PMCID: PMC10669475 DOI: 10.3390/biom13111621] [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: 09/21/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Zebrafish are increasingly becoming an important model organism for studying the pathophysiological mechanisms of human diseases and investigating how these mechanisms can be effectively targeted using compounds that may open avenues to novel treatments for patients. The zebrafish skeleton has been particularly instrumental in modeling bone diseases as-contrary to other model organisms-the lower load on the skeleton of an aquatic animal enables mutants to survive to early adulthood. In this respect, the axial skeletons of zebrafish have been a good read-out for congenital spinal deformities such as scoliosis and degenerative disorders such as osteoporosis and osteoarthritis, in which aberrant mineralization in humans is reflected in the respective zebrafish models. Interestingly, there have been several reports of hereditary multisystemic diseases that do not affect the vertebral column in human patients, while the corresponding zebrafish models systematically show anomalies in mineralization and morphology of the spine as their leading or, in some cases, only phenotype. In this review, we describe such examples, highlighting the underlying mechanisms, the already-used or potential power of these models to help us understand and amend the mineralization process, and the outstanding questions on how and why this specific axial type of aberrant mineralization occurs in these disease models.
Collapse
Affiliation(s)
- Judith Van Wynsberghe
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Ectopic Mineralization Research Group, 9000 Ghent, Belgium
| | - Olivier M Vanakker
- Center for Medical Genetics, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
- Ectopic Mineralization Research Group, 9000 Ghent, Belgium
| |
Collapse
|
9
|
Fan YL, Hsu FR, Wang Y, Liao LD. Unlocking the Potential of Zebrafish Research with Artificial Intelligence: Advancements in Tracking, Processing, and Visualization. Med Biol Eng Comput 2023; 61:2797-2814. [PMID: 37558927 DOI: 10.1007/s11517-023-02903-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Zebrafish have become a widely accepted model organism for biomedical research due to their strong cortisol stress response, behavioral strain differences, and sensitivity to both drug treatments and predators. However, experimental zebrafish studies generate substantial data that must be analyzed through objective, accurate, and repeatable analysis methods. Recently, advancements in artificial intelligence (AI) have enabled automated tracking, image recognition, and data analysis, leading to more efficient and insightful investigations. In this review, we examine key AI applications in zebrafish research, including behavior analysis, genomics, and neuroscience. With the development of deep learning technology, AI algorithms have been used to precisely analyze and identify images of zebrafish, enabling automated testing and analysis. By applying AI algorithms in genomics research, researchers have elucidated the relationship between genes and biology, providing a better basis for the development of disease treatments and gene therapies. Additionally, the development of more effective neuroscience tools could help researchers better understand the complex neural networks in the zebrafish brain. In the future, further advancements in AI technology are expected to enable more extensive and in-depth medical research applications in zebrafish, improving our understanding of this important animal model. This review highlights the potential of AI technology in achieving the full potential of zebrafish research by enabling researchers to efficiently track, process, and visualize the outcomes of their experiments.
Collapse
Affiliation(s)
- Yi-Ling Fan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan
- Department of Information Engineering and Computer Science, Feng Chia University, Taichung, 407, Taiwan
| | - Fang-Rong Hsu
- Department of Information Engineering and Computer Science, Feng Chia University, Taichung, 407, Taiwan
| | - Yuhling Wang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan
- Department of Electrical Engineering, National United University, 2, Lien-Da, Nan-Shih Li, Miaoli, 360302, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35, Keyan Road, Zhunan Town, Miaoli County, 35053, Taiwan.
| |
Collapse
|
10
|
Maslov GO, Zabegalov KN, Demin KA, Kolesnikova TO, Kositsyn YM, de Abreu MS, Petersen EV, Kalueff AV. Towards experimental models of delirium utilizing zebrafish. Behav Brain Res 2023; 453:114607. [PMID: 37524203 DOI: 10.1016/j.bbr.2023.114607] [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: 05/25/2023] [Revised: 07/28/2023] [Accepted: 07/29/2023] [Indexed: 08/02/2023]
Abstract
Delirium is an acute neuropsychiatric condition characterized by impaired behavior and cognition. Although the syndrome has been known for millennia, its CNS mechanisms and risk factors remain poorly understood. Experimental animal models, especially rodent-based, are commonly used to probe various pathogenetic aspects of delirium. Complementing rodents, the zebrafish (Danio rerio) emerges as a promising novel model organism to study delirium. Zebrafish demonstrate high genetic and physiological homology to mammals, easy maintenance, robust behaviors in various sensitive behavioral tests, and the potential to screen for pharmacological agents relevant to delirium. Here, we critically discuss recent developments in the field, and emphasize the developing utility of zebrafish models for translational studies of delirium and deliriant drugs. Overall, the zebrafish represents a valuable and promising aquatic model species whose use may help understand delirium etiology, as well as develop novel therapies for this severely debilitating disorder.
Collapse
Affiliation(s)
- Gleb O Maslov
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; Ural Federal University, Ekaterinburg, Russia
| | | | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Tatiana O Kolesnikova
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Yuriy M Kositsyn
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Murilo S de Abreu
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia.
| | - Elena V Petersen
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russia
| | - Allan V Kalueff
- Neurobiology Program, Sirius University of Science and Technology, Sochi, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia; Novosibirsk State University, Novosibirsk, Russia; Laboratory of Preclinical Bioscreening, Granov Russian Research Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, Pesochny, Russia; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
11
|
Henke K, Farmer DT, Niu X, Kraus JM, Galloway JL, Youngstrom DW. Genetically engineered zebrafish as models of skeletal development and regeneration. Bone 2023; 167:116611. [PMID: 36395960 PMCID: PMC11080330 DOI: 10.1016/j.bone.2022.116611] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
Zebrafish (Danio rerio) are aquatic vertebrates with significant homology to their terrestrial counterparts. While zebrafish have a centuries-long track record in developmental and regenerative biology, their utility has grown exponentially with the onset of modern genetics. This is exemplified in studies focused on skeletal development and repair. Herein, the numerous contributions of zebrafish to our understanding of the basic science of cartilage, bone, tendon/ligament, and other skeletal tissues are described, with a particular focus on applications to development and regeneration. We summarize the genetic strengths that have made the zebrafish a powerful model to understand skeletal biology. We also highlight the large body of existing tools and techniques available to understand skeletal development and repair in the zebrafish and introduce emerging methods that will aid in novel discoveries in skeletal biology. Finally, we review the unique contributions of zebrafish to our understanding of regeneration and highlight diverse routes of repair in different contexts of injury. We conclude that zebrafish will continue to fill a niche of increasing breadth and depth in the study of basic cellular mechanisms of skeletal biology.
Collapse
Affiliation(s)
- Katrin Henke
- Department of Orthopaedics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - D'Juan T Farmer
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095, USA.
| | - Xubo Niu
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Jessica M Kraus
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
12
|
Mrinalini R, Tamilanban T, Naveen Kumar V, Manasa K. Zebrafish - The Neurobehavioural Model in Trend. Neuroscience 2022; 520:95-118. [PMID: 36549602 DOI: 10.1016/j.neuroscience.2022.12.016] [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: 03/24/2022] [Revised: 12/11/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Zebrafish (Danio rerio) is currently in vogue as a prevalently used experimental model for studies concerning neurobehavioural disorders and associated fields. Since the 1960s, this model has succeeded in breaking most barriers faced in the hunt for an experimental model. From its appearance to its high parity with human beings genetically, this model renders itself as an advantageous experimental lab animal. Neurobehavioural disorders have always posed an arduous task in terms of their detection as well as in determining their exact etiology. They are still, in most cases, diseases of interest for inventing or discovering novel pharmacological interventions. Thus, the need for a harbinger experimental model for studying neurobehaviours is escalating. Ensuring the same model is used for studying several neuro-studies conserves the results from inter-species variations. For this, we need a model that satisfies all the pre-requisite conditions to be made the final choice of model for neurobehavioural studies. This review recapitulates the progress of zebrafish as an experimental model with its most up-to-the-minute advances in the area. Various tests, assays, and responses employed using zebrafish in screening neuroactive drugs have been tabulated effectively. The tools, techniques, protocols, and apparatuses that bolster zebrafish studies are discussed. The probable research that can be done using zebrafish has also been briefly outlined. The various breeding and maintenance methods employed, along with the information on various strains available and most commonly used, are also elaborated upon, supplementing Zebrafish's use in neuroscience.
Collapse
Affiliation(s)
- R Mrinalini
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur, India - 603203
| | - T Tamilanban
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur, India - 603203
| | - V Naveen Kumar
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur, India - 603203.
| | - K Manasa
- Department of Pharmacology, SRM College of Pharmacy, SRMIST, Kattankulathur, India - 603203
| |
Collapse
|
13
|
Ohashi ASC, de Souza Schacher HR, Pizzato CS, Vianna MRMR, de Menezes LM. Zebrafish as model for studies in dentistry. J Orthod Sci 2022; 11:46. [PMID: 36411806 PMCID: PMC9674940 DOI: 10.4103/jos.jos_41_22] [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: 05/13/2022] [Revised: 07/22/2022] [Accepted: 08/02/2022] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Over the last years, zebrafish has gained prominence in the biomedical community. It is currently considered one of the best vertebrate animal models for various types of studies, such as toxicology and developmental biology. OBJECTIVE The aim of this study was to conduct a literature review on the use of zebrafish in dentistry and whether this animal model could be a viable alternative for performing different types of studies in this area. METHODS A literature search was performed using the PubMed, Lilacs, Embase, and Dentistry and Oral Sciences Source. The keywords used as search terms were zebrafish and dentistry. The selection criteria were articles published in English that used zebrafish as an animal model in dentistry, oral health, and craniofacial growth/development. RESULTS The electronic search of literature yielded 421 articles. After the analysis of the abstracts, 29 articles were selected for an in-depth analysis and reading of the full text. CONCLUSIONS All studies included in this review confirm zebrafish's excellence as an animal model for various types of dentistry studies, as well as assisting and complementing other studies involving mammals.
Collapse
Affiliation(s)
- Amanda S. C. Ohashi
- PhD Students, Dental Program, School of Health and Life Sciences, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Helena R. de Souza Schacher
- PhD Students, Dental Program, School of Health and Life Sciences, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Mônica R. M. R. Vianna
- Professor at the Postgraduate Programs in Biology Cellular and Molecular and in Ecology and Evolution of Biodiversity, ZebLab, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Luciane M. de Menezes
- Professor at the School of Health Sciences and life (Dental Program) of the Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil,Address for correspondence: Dr. Luciane M. de Menezes, Dental Program, School of Health and Life Sciences, Pontifícia Universidade Católica do Rio Grande do Sul, 6681 Ipiranga Avenue, Building n. 6, Porto Alegre, RS, 90619-900, Brazil. E-mail:
| |
Collapse
|
14
|
Disease Modeling of Rare Neurological Disorders in Zebrafish. Int J Mol Sci 2022; 23:ijms23073946. [PMID: 35409306 PMCID: PMC9000079 DOI: 10.3390/ijms23073946] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/31/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023] Open
Abstract
Rare diseases are those which affect a small number of people compared to the general population. However, many patients with a rare disease remain undiagnosed, and a large majority of rare diseases still have no form of viable treatment. Approximately 40% of rare diseases include neurologic and neurodevelopmental disorders. In order to understand the characteristics of rare neurological disorders and identify causative genes, various model organisms have been utilized extensively. In this review, the characteristics of model organisms, such as roundworms, fruit flies, and zebrafish, are examined, with an emphasis on zebrafish disease modeling in rare neurological disorders.
Collapse
|
15
|
Alov P, Al Sharif M, Aluani D, Chegaev K, Dinic J, Divac Rankov A, Fernandes MX, Fusi F, García-Sosa AT, Juvonen R, Kondeva-Burdina M, Padrón JM, Pajeva I, Pencheva T, Puerta A, Raunio H, Riganti C, Tsakovska I, Tzankova V, Yordanov Y, Saponara S. A Comprehensive Evaluation of Sdox, a Promising H2S-Releasing Doxorubicin for the Treatment of Chemoresistant Tumors. Front Pharmacol 2022; 13:831791. [PMID: 35321325 PMCID: PMC8936434 DOI: 10.3389/fphar.2022.831791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Sdox is a hydrogen sulfide (H2S)-releasing doxorubicin effective in P-glycoprotein-overexpressing/doxorubicin-resistant tumor models and not cytotoxic, as the parental drug, in H9c2 cardiomyocytes. The aim of this study was the assessment of Sdox drug-like features and its absorption, distribution, metabolism, and excretion (ADME)/toxicity properties, by a multi- and transdisciplinary in silico, in vitro, and in vivo approach. Doxorubicin was used as the reference compound. The in silico profiling suggested that Sdox possesses higher lipophilicity and lower solubility compared to doxorubicin, and the off-targets prediction revealed relevant differences between Dox and Sdox towards several cancer targets, suggesting different toxicological profiles. In vitro data showed that Sdox is a substrate with lower affinity for P-glycoprotein, less hepatotoxic, and causes less oxidative damage than doxorubicin. Both anthracyclines inhibited CYP3A4, but not hERG currents. Unlike doxorubicin, the percentage of zebrafish live embryos at 72 hpf was not affected by Sdox treatment. In conclusion, these findings demonstrate that Sdox displays a more favorable drug-like ADME/toxicity profile than doxorubicin, different selectivity towards cancer targets, along with a greater preclinical efficacy in resistant tumors. Therefore, Sdox represents a prototype of innovative anthracyclines, worthy of further investigations in clinical settings.
Collapse
Affiliation(s)
- Petko Alov
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Merilin Al Sharif
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Denitsa Aluani
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Jelena Dinic
- Department of Neurobiology, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Divac Rankov
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Miguel X. Fernandes
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Risto Juvonen
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Magdalena Kondeva-Burdina
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - José M. Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Ilza Pajeva
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tania Pencheva
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, Spain
| | - Hannu Raunio
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | - Ivanka Tsakovska
- Department of QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Virginia Tzankova
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Yordan Yordanov
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia, Bulgaria
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Siena, Italy
- *Correspondence: Simona Saponara,
| |
Collapse
|
16
|
Abozaid A, Hung J, Tsang B, Motlana K, Al-Ani R, Gerlai R. Behavioral effects of acute ethanol in larval zebrafish (D. rerio) depend on genotype and volume of experimental well. Prog Neuropsychopharmacol Biol Psychiatry 2022; 112:110411. [PMID: 34363865 DOI: 10.1016/j.pnpbp.2021.110411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 01/10/2023]
Abstract
Ethanol consumption is a worldwide problem. Sensitivity to acute effects of ethanol influences the development of chronic ethanol abuse and ethanol dependence. Environmental and genetic factors have been found to contribute to differential effects of acute ethanol. Animal models have been employed to investigate these factors. An increasingly frequently utilized animal model in ethanol research is the zebrafish. A large proportion of ethanol studies with zebrafish have been conducted with adult zebrafish. However, high throughput drug and mutation screens are particularly well adapted to larval zebrafish. These studies are often carried out using the 96-well-plate that allows monitoring large numbers of fish efficiently. Here, we investigate the effects of acute (30 min long) ethanol exposure in 8-day post-fertilization (dpf) old zebrafish. We compare four genetically distinct populations (strains) of zebrafish, measuring numerous parameters of their swim path in two well sizes, i.e., in the 96-well-plate (small volume wells) and in the 6-well-plate (large volume wells). In general, we found that the highest dose of ethanol (1% vol/vol) reduced swim speed, increased duration of immobility, increased turn angle, and increased intra-individual variance of turn angle, while the intermediate dose (0.5%) had a less strong effect, compared to control. However, we also found that these ethanol effects were strain dependent and, in general, were better detected in the larger volume well. We conclude that larval zebrafish are appropriate for quantification of acute ethanol effects and also for the analysis of environmental and genetic factors that influence these effects. We also speculate that using larger wells will likely increase sensitivity of detection and precision in screening applications.
Collapse
Affiliation(s)
- Amira Abozaid
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Joshua Hung
- Department of Biology, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Benjamin Tsang
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada; Department of Critical Care Medicine, Hospital for Sick Children, Toronto, Canada; Institute of Psychiatry, Psychology & Neuroscience, Kings College London, London, United Kingdom
| | - Keza Motlana
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Reem Al-Ani
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Robert Gerlai
- Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada; Department of Cell and Systems Biology, University of Toronto, Toronto, Canada.
| |
Collapse
|
17
|
Messina A, Potrich D, Schiona I, Sovrano VA, Vallortigara G. The Sense of Number in Fish, with Particular Reference to Its Neurobiological Bases. Animals (Basel) 2021; 11:ani11113072. [PMID: 34827804 PMCID: PMC8614421 DOI: 10.3390/ani11113072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/13/2021] [Accepted: 10/22/2021] [Indexed: 01/29/2023] Open
Abstract
Simple Summary The ability to deal with quantity, both discrete (numerosities) and continuous (spatial or temporal extent) developed from an evolutionarily conserved system for approximating numerical magnitude. Non-symbolic number cognition based on an approximate sense of magnitude has been documented in a variety of vertebrate species, including fish. Fish, in particular zebrafish, are widely used as models for the investigation of the genetics and molecular mechanisms of behavior, and thus may be instrumental to development of a neurobiology of number cognition. We review here the behavioural studies that have permitted to identify numerical abilities in fish, and the current status of the research related to the neurobiological bases of these abilities with special reference to zebrafish. Combining behavioural tasks with molecular genetics, molecular biology and confocal microscopy, a role of the retina and optic tectum in the encoding of continuous magnitude in larval zebrafish has been reported, while the thalamus and the dorso-central subdivision of pallium in the encoding of discrete magnitude (number) has been documented in adult zebrafish. Research in fish, in particular zebrafish, may reveal instrumental for identifying and characterizing the molecular signature of neurons involved in quantity discrimination processes of all vertebrates, including humans. Abstract It is widely acknowledged that vertebrates can discriminate non-symbolic numerosity using an evolutionarily conserved system dubbed Approximate Number System (ANS). Two main approaches have been used to assess behaviourally numerosity in fish: spontaneous choice tests and operant training procedures. In the first, animals spontaneously choose between sets of biologically-relevant stimuli (e.g., conspecifics, food) differing in quantities (smaller or larger). In the second, animals are trained to associate a numerosity with a reward. Although the ability of fish to discriminate numerosity has been widely documented with these methods, the molecular bases of quantities estimation and ANS are largely unknown. Recently, we combined behavioral tasks with molecular biology assays (e.g c-fos and egr1 and other early genes expression) showing that the thalamus and the caudal region of dorso-central part of the telencephalon seem to be activated upon change in numerousness in visual stimuli. In contrast, the retina and the optic tectum mainly responded to changes in continuous magnitude such as stimulus size. We here provide a review and synthesis of these findings.
Collapse
Affiliation(s)
- Andrea Messina
- Centre for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (D.P.); (I.S.); (V.A.S.)
- Correspondence: (A.M.); (G.V.)
| | - Davide Potrich
- Centre for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (D.P.); (I.S.); (V.A.S.)
| | - Ilaria Schiona
- Centre for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (D.P.); (I.S.); (V.A.S.)
| | - Valeria Anna Sovrano
- Centre for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (D.P.); (I.S.); (V.A.S.)
- Department of Psychology and Cognitive Science, University of Trento, 38068 Rovereto, Italy
| | - Giorgio Vallortigara
- Centre for Mind/Brain Sciences, University of Trento, 38068 Rovereto, Italy; (D.P.); (I.S.); (V.A.S.)
- Correspondence: (A.M.); (G.V.)
| |
Collapse
|
18
|
Vroman R, Malfait AM, Miller RE, Malfait F, Syx D. Animal Models of Ehlers-Danlos Syndromes: Phenotype, Pathogenesis, and Translational Potential. Front Genet 2021; 12:726474. [PMID: 34712265 PMCID: PMC8547655 DOI: 10.3389/fgene.2021.726474] [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: 06/16/2021] [Accepted: 08/10/2021] [Indexed: 01/09/2023] Open
Abstract
The Ehlers-Danlos syndromes (EDS) are a group of heritable connective tissues disorders mainly characterized by skin hyperextensibility, joint hypermobility and generalized tissue fragility. Currently, 14 EDS subtypes each with particular phenotypic features are recognized and are caused by genetic defects in 20 different genes. All of these genes are involved in the biosynthesis and/or fibrillogenesis of collagens at some level. Although great progress has been made in elucidating the molecular basis of different EDS subtypes, the pathogenic mechanisms underlying the observed phenotypes remain poorly understood, and consequentially, adequate treatment and management options for these conditions remain scarce. To date, several animal models, mainly mice and zebrafish, have been described with defects in 14 of the 20 hitherto known EDS-associated genes. These models have been instrumental in discerning the functions and roles of the corresponding proteins during development, maturation and repair and in portraying their roles during collagen biosynthesis and/or fibrillogenesis, for some even before their contribution to an EDS phenotype was elucidated. Additionally, extensive phenotypical characterization of these models has shown that they largely phenocopy their human counterparts, with recapitulation of several clinical hallmarks of the corresponding EDS subtype, including dermatological, cardiovascular, musculoskeletal and ocular features, as well as biomechanical and ultrastructural similarities in tissues. In this narrative review, we provide a comprehensive overview of animal models manifesting phenotypes that mimic EDS with a focus on engineered mouse and zebrafish models, and their relevance in past and future EDS research. Additionally, we briefly discuss domestic animals with naturally occurring EDS phenotypes. Collectively, these animal models have only started to reveal glimpses into the pathophysiological aspects associated with EDS and will undoubtably continue to play critical roles in EDS research due to their tremendous potential for pinpointing (common) signaling pathways, unveiling possible therapeutic targets and providing opportunities for preclinical therapeutic interventions.
Collapse
Affiliation(s)
- Robin Vroman
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Anne-Marie Malfait
- Division of Rheumatology, Rush University Medical Center, Chicago, IL, United States
| | - Rachel E. Miller
- Division of Rheumatology, Rush University Medical Center, Chicago, IL, United States
| | - Fransiska Malfait
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Delfien Syx
- Center for Medical Genetics, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
19
|
Wang Z, Ding Y, Satta S, Roustaei M, Fei P, Hsiai TK. A hybrid of light-field and light-sheet imaging to study myocardial function and intracardiac blood flow during zebrafish development. PLoS Comput Biol 2021; 17:e1009175. [PMID: 34228702 PMCID: PMC8284633 DOI: 10.1371/journal.pcbi.1009175] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 07/16/2021] [Accepted: 06/11/2021] [Indexed: 01/07/2023] Open
Abstract
Biomechanical forces intimately contribute to cardiac morphogenesis. However, volumetric imaging to investigate the cardiac mechanics with high temporal and spatial resolution remains an imaging challenge. We hereby integrated light-field microscopy (LFM) with light-sheet fluorescence microscopy (LSFM), coupled with a retrospective gating method, to simultaneously access myocardial contraction and intracardiac blood flow at 200 volumes per second. While LSFM allows for the reconstruction of the myocardial function, LFM enables instantaneous acquisition of the intracardiac blood cells traversing across the valves. We further adopted deformable image registration to quantify the ventricular wall displacement and particle tracking velocimetry to monitor intracardiac blood flow. The integration of LFM and LSFM enabled the time-dependent tracking of the individual blood cells and the differential rates of segmental wall displacement during a cardiac cycle. Taken together, we demonstrated a hybrid system, coupled with our image analysis pipeline, to simultaneously capture the myocardial wall motion with intracardiac blood flow during cardiac development. During the conception of the heart, cardiac muscular contraction and blood flow generate biomechanical forces to influence the functional and structural development. To elucidate the underlying biomechanical mechanisms, we have embraced the zebrafish system for the ease of genetic and pharmacological manipulations and its rapidity for organ development. However, acquiring the dynamic processes (space + time domain) in the small beating zebrafish heart remains a challenge. In the presence of a rapid heartbeat, microscopy is confined by temporal resolution to image the cardiac contraction and blood flow. In this context, we demonstrated an integrated light-sheet and light-field imaging system to visualize cardiac contraction along with the flowing blood cells inside the cardiac chambers. Assuming the periodicity of the cardiac cycle, we synchronized the image data in post-processing for 3-D reconstruction. We further quantified the velocity of the various regions of cardiac muscular contraction, and tracked the individual blood cells during the cardiac cycles. The time-dependent velocity maps allow for uncovering differential segments of cardiac contraction and relaxation, and for revealing the patterns of blood flow. Thus, our integrated light-sheet and light-field imaging system provides an experimental basis to further investigate cardiac function and development.
Collapse
Affiliation(s)
- Zhaoqiang Wang
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
| | - Yichen Ding
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
| | - Sandro Satta
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
| | - Mehrdad Roustaei
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
| | - Peng Fei
- School of Optical and Electronic Information-Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (PF); (TKH)
| | - Tzung K. Hsiai
- Department of Bioengineering, University of California, Los Angeles, California, United States of America
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, California, United States of America
- Department of Medicine, Greater Los Angeles VA Healthcare System, Los Angeles, California, United States of America
- * E-mail: (PF); (TKH)
| |
Collapse
|
20
|
Sheets L, Holmgren M, Kindt KS. How Zebrafish Can Drive the Future of Genetic-based Hearing and Balance Research. J Assoc Res Otolaryngol 2021; 22:215-235. [PMID: 33909162 PMCID: PMC8110678 DOI: 10.1007/s10162-021-00798-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Over the last several decades, studies in humans and animal models have successfully identified numerous molecules required for hearing and balance. Many of these studies relied on unbiased forward genetic screens based on behavior or morphology to identify these molecules. Alongside forward genetic screens, reverse genetics has further driven the exploration of candidate molecules. This review provides an overview of the genetic studies that have established zebrafish as a genetic model for hearing and balance research. Further, we discuss how the unique advantages of zebrafish can be leveraged in future genetic studies. We explore strategies to design novel forward genetic screens based on morphological alterations using transgenic lines or behavioral changes following mechanical or acoustic damage. We also outline how recent advances in CRISPR-Cas9 can be applied to perform reverse genetic screens to validate large sequencing datasets. Overall, this review describes how future genetic studies in zebrafish can continue to advance our understanding of inherited and acquired hearing and balance disorders.
Collapse
Affiliation(s)
- Lavinia Sheets
- Department of Otolaryngology-Head & Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Melanie Holmgren
- Department of Otolaryngology-Head & Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Katie S Kindt
- Section On Sensory Cell Development and Function, National Institutes On Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, USA.
| |
Collapse
|
21
|
Rojas AM, Shiau CE. Brain-localized and Intravenous Microinjections in the Larval Zebrafish to Assess Innate Immune Response. Bio Protoc 2021; 11:e3978. [PMID: 33889672 DOI: 10.21769/bioprotoc.3978] [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: 11/18/2020] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 11/02/2022] Open
Abstract
Creating a robust and controlled infection model is imperative for studying the innate immune response. Leveraging the particular strengths of the zebrafish model system, such as optical transparency, ex utero development, and large clutch size, allows for the development of methods that yield consistent and reproducible results. We created a robust model for activation of innate immunity by microinjecting bacterial particles or live bacteria into larval zebrafish, unlike previous studies which largely restricted such manipulations to embryonic stages of zebrafish. The ability to introduce stimuli locally or systemically at larval stages provides significant advantages to examine host response in more mature tissues as well as the possibility to interrogate adaptive immunity at older larval stages. This protocol describes two distinct modes of microinjection to introduce lipopolysaccharide (LPS) or bacteria into the living larval zebrafish: one localized to the brain, and another into the bloodstream via the caudal vein plexus. Graphic abstract: Schematic shows the two distinct modes of larval zebrafish microinjection, either in the brain parenchyma or in the blood stream intravenously. Reagents introduced into the zebrafish to assess immune response are depicted in the "injection components" as described in the protocol.
Collapse
Affiliation(s)
- Alison M Rojas
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Celia E Shiau
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
22
|
Costa FV, Rosa LV, Quadros VA, de Abreu MS, Santos ARS, Sneddon LU, Kalueff AV, Rosemberg DB. The use of zebrafish as a non-traditional model organism in translational pain research: the knowns and the unknowns. Curr Neuropharmacol 2021; 20:476-493. [PMID: 33719974 DOI: 10.2174/1570159x19666210311104408] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/24/2021] [Accepted: 02/28/2021] [Indexed: 11/22/2022] Open
Abstract
The ability of the nervous system to detect a wide range of noxious stimuli is crucial to avoid life-threatening injury and to trigger protective behavioral and physiological responses. Pain represents a complex phenomenon, including nociception associated with cognitive and emotional processing. Animal experimental models have been developed to understand the mechanisms involved in pain response, as well as to discover novel pharmacological and non-pharmacological anti-pain therapies. Due to the genetic tractability, similar physiology, low cost, and rich behavioral repertoire, the zebrafish (Danio rerio) has been considered a powerful aquatic model for modeling pain responses. Here, we summarize the molecular machinery of zebrafish to recognize painful stimuli, as well as emphasize how zebrafish-based pain models have been successfully used to understand specific molecular, physiological, and behavioral changes following different algogens and/or noxious stimuli (e.g., acetic acid, formalin, histamine, Complete Freund's Adjuvant, cinnamaldehyde, allyl isothiocyanate, and fin clipping). We also discuss recent advances in zebrafish-based studies and outline the potential advantages and limitations of the existing models to examine the mechanisms underlying pain responses from an evolutionary and translational perspective. Finally, we outline how zebrafish models can represent emergent tools to explore pain behaviors and pain-related mood disorders, as well as to facilitate analgesic therapy screening in translational pain research.
Collapse
Affiliation(s)
- Fabiano V Costa
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria RS. Brazil
| | - Luiz V Rosa
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria RS. Brazil
| | - Vanessa A Quadros
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria RS. Brazil
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo (UPF), Passo Fundo, RS. Brazil
| | - Adair R S Santos
- Laboratory of Neurobiology of Pain and Inflammation, Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Trindade, Florianópolis, SC. Brazil
| | - Lynne U Sneddon
- University of Gothenburg, Department of Biological & Environmental Sciences, Box 461, SE-405 30 Gothenburg. Sweden
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg. Russian Federation
| | - Denis B Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria RS. Brazil
| |
Collapse
|
23
|
Zebrafish as an animal model for biomedical research. Exp Mol Med 2021; 53:310-317. [PMID: 33649498 PMCID: PMC8080808 DOI: 10.1038/s12276-021-00571-5] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 12/15/2022] Open
Abstract
Zebrafish have several advantages compared to other vertebrate models used in modeling human diseases, particularly for large-scale genetic mutant and therapeutic compound screenings, and other biomedical research applications. With the impactful developments of CRISPR and next-generation sequencing technology, disease modeling in zebrafish is accelerating the understanding of the molecular mechanisms of human genetic diseases. These efforts are fundamental for the future of precision medicine because they provide new diagnostic and therapeutic solutions. This review focuses on zebrafish disease models for biomedical research, mainly in developmental disorders, mental disorders, and metabolic diseases.
Collapse
|
24
|
Usui Y, Watanabe M. Role of the Connexin C-terminus in skin pattern formation of Zebrafish. BBA ADVANCES 2021; 1:100006. [PMID: 37082017 PMCID: PMC10074918 DOI: 10.1016/j.bbadva.2021.100006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background Zebrafish display a striped skin pattern on their body; two types of connexins, namely, Connexin39.4 (Cx39.4) and Connexin41.8 (Cx41.8), are involved in stripe pattern formation. Herein, we investigated the role of the C-terminal (CT) domains of Cx39.4 and Cx41.8 in vivo and in vitro. Methods To investigate the role of CT domains in vivo, we established transgenic zebrafish lines expressing the CT-domain-modified connexin series in pigmented cells and observed skin patterns in fish. To investigate the role of the CT domains in vitro, we expressed the CT-domain modified connexin series in Neuro-2a (N2a) cells and calculated the plaque formation frequency. Results The overexpression of Cx39.4 lacking a CT domain produced skin patterns similar to that produced by full-length Cx39.4 in the cx39.4 -/- mutant and in cx39.4 and cx41.8 double-knockout mutant zebrafish. Fluorescence-protein-fused CT-domain-modified Cx39.4 formed gap junction plaques between N2a cells. The overexpression of CT-truncated Cx41.8 rescued the mutant phenotype in the cx41.8 -/- mutant but did not function in the double knockout zebrafish. Fluorescence-protein-fused CT-truncated Cx41.8 hardly formed plaques between N2a cells without Cx39.4 but formed gap junction plaques when co-expressed with Cx39.4. Conclusions The CT domain of Cx39.4 is not required for protein function, at least in the pigment cells of zebrafish. However, the need for the CT domain of Cx41.8 depends on Cx39.4 expression. General significance These results provide evidence for the interactions between Cx39.4 and Cx41.8 in pigment cells of zebrafish and suggest that at least one connexin must have a CT domain.
Collapse
|
25
|
Carroll SH, Macias Trevino C, Li EB, Kawasaki K, Myers N, Hallett SA, Alhazmi N, Cotney J, Carstens RP, Liao EC. An Irf6- Esrp1/2 regulatory axis controls midface morphogenesis in vertebrates. Development 2020; 147:dev194498. [PMID: 33234718 PMCID: PMC7774891 DOI: 10.1242/dev.194498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022]
Abstract
Irf6 and Esrp1 are important for palate development across vertebrates. In zebrafish, we found that irf6 regulates the expression of esrp1 We detailed overlapping Irf6 and Esrp1/2 expression in mouse orofacial epithelium. In zebrafish, irf6 and esrp1/2 share expression in periderm, frontonasal ectoderm and oral epithelium. Genetic disruption of irf6 and esrp1/2 in zebrafish resulted in cleft of the anterior neurocranium. The esrp1/2 mutant also developed cleft of the mouth opening. Lineage tracing of cranial neural crest cells revealed that the cleft resulted not from migration defect, but from impaired chondrogenesis. Analysis of aberrant cells within the cleft revealed expression of sox10, col1a1 and irf6, and these cells were adjacent to krt4+ and krt5+ cells. Breeding of mouse Irf6; Esrp1; Esrp2 compound mutants suggested genetic interaction, as the triple homozygote and the Irf6; Esrp1 double homozygote were not observed. Further, Irf6 heterozygosity reduced Esrp1/2 cleft severity. These studies highlight the complementary analysis of Irf6 and Esrp1/2 in mouse and zebrafish, and identify a unique aberrant cell population in zebrafish expressing sox10, col1a1 and irf6 Future work characterizing this cell population will yield additional insight into cleft pathogenesis.
Collapse
Affiliation(s)
- Shannon H. Carroll
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Claudio Macias Trevino
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| | | | - Kenta Kawasaki
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
| | - Nikita Myers
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Shawn A. Hallett
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nora Alhazmi
- Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Justin Cotney
- Department of Genetics and Genome Sciences, University of Connecticut Health, CT 06030, USA
| | - Russ P. Carstens
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric C. Liao
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Shriners Hospital for Children, Boston, MA 02114, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Chen H, Shan G. The physiological function of long-noncoding RNAs. Noncoding RNA Res 2020; 5:178-184. [PMID: 32959025 PMCID: PMC7494506 DOI: 10.1016/j.ncrna.2020.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
The physiological processes of cells and organisms are regulated by various biological macromolecules, including long-noncoding RNAs (lncRNAs), which cannot be translated into protein and are different from small-noncoding RNAs on their length. In animals, lncRNAs are involved in development, metabolism, reproduction, aging and other life events by cis or trans effects. For many functional lncRNAs, there is growing evidence that they play different roles on cellular level and organismal level. On the other hand, many annotated lncRNAs are not essential and could be transcription noises. In this minireview, we investigate the physiological function of lncRNAs in cells and focus on their functions and functional mechanisms on the organismal level. The studies on lncRNAs using different classic animal models such as worms and flies are summarized and discussed in this article.
Collapse
Affiliation(s)
- He Chen
- CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Center for Excellence in Molecular Cell Science, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui Province, 230027, China
| | - Ge Shan
- CAS Key Laboratory of Innate Immunity and Chronic Disease, CAS Center for Excellence in Molecular Cell Science, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui Province, 230027, China
| |
Collapse
|
27
|
Schulte C, Nagel R. Testing Acute Toxicity in the Embryo of Zebrafish, Brachydanio rerio, as an Alternative to the Acute Fish Test: Preliminary Results. Altern Lab Anim 2020. [DOI: 10.1177/026119299402200104] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Acute toxicity testing in fish is a standard method used in estimating the influences of chemicals on aquatic vertebrates. The ecotoxicological data obtained from acute toxicity tests in fish are, however, not sufficiently reliable to justify the continued use of this test. Fertilised eggs of zebrafish ( Brachydanio rerio) were used to test the acute toxicity of chemicals. They were chosen because the development of B. rerio has been studied extensively and information already exists concerning the normal development of this species. The following parameters of the development of B. rerio were observed: coagulation of the egg, gastrulation, number of somites, movement, development of organs, pigmentation, heartbeat and circulation. Some of these toxicological endpoints indicate lethality. In addition, various interactions between the test chemical and the embryos can be measured by investigating these parameters. Six chemicals (2,4-dinitrophenol, 4,6-dinitro-o-cresol, malathion, carbaryl, phenol and 4-nitrophenol) were selected for testing in the embryos of zebrafish within the first 48 hours of their development. The toxicities of the test chemicals to zebrafish embryos were compared with their acute toxicities to adult fish. Further investigations with more chemicals are in progress. The effective concentrations (EC50) and the lowest effect concentrations (LOEC) investigated in this preliminary study were comparable to the LC50 values for adult fish. The use of this test system as an alternative method to testing toxicity in adult fish is discussed.
Collapse
Affiliation(s)
- Christoph Schulte
- Department of Zoology, Johannes Gutenberg University, Saarstrasse 21, D-55099 Mainz, Germany
| | - Roland Nagel
- Department of Zoology, Johannes Gutenberg University, Saarstrasse 21, D-55099 Mainz, Germany
| |
Collapse
|
28
|
de Abreu MS, C V V Giacomini A, Genario R, Fontana BD, Parker MO, Marcon L, Scolari N, Bueno B, Demin KA, Galstyan D, Kolesnikova TO, Amstislavskaya TG, Zabegalov KN, Strekalova T, Kalueff AV. Zebrafish models of impulsivity and impulse control disorders. Eur J Neurosci 2020; 52:4233-4248. [PMID: 32619029 DOI: 10.1111/ejn.14893] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/25/2020] [Accepted: 06/18/2020] [Indexed: 12/22/2022]
Abstract
Impulse control disorders (ICDs) are characterized by generalized difficulty controlling emotions and behaviors. ICDs are a broad group of the central nervous system (CNS) disorders including conduct disorder, intermittent explosive, oppositional-defiant disorder, antisocial personality disorder, kleptomania, pyromania and other illnesses. Although they all share a common feature (aberrant impulsivity), their pathobiology is complex and poorly understood. There are also currently no ICD-specific therapies to treat these illnesses. Animal models are a valuable tool for studying ICD pathobiology and potential therapies. The zebrafish (Danio rerio) has become a useful model organism to study CNS disorders due to high genetic and physiological homology to mammals, and sensitivity to various pharmacological and genetic manipulations. Here, we summarize experimental models of impulsivity and ICD in zebrafish and highlight their growing translational significance. We also emphasize the need for further development of zebrafish ICD models to improve our understanding of their pathogenesis and to search for novel therapeutic treatments.
Collapse
Affiliation(s)
- Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil.,The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, USA
| | - Ana C V V Giacomini
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil.,Postgraduate Program in Environmental Sciences, University of Passo Fundo, Passo Fundo, Brazil
| | - Rafael Genario
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
| | - Barbara D Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK
| | - Matthew O Parker
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, UK
| | - Leticia Marcon
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
| | - Naiara Scolari
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
| | - Barbara Bueno
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia.,Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - David Galstyan
- Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia
| | - Tatyana O Kolesnikova
- Institute of Experimental Medicine, Almazov National Medical Research Center, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia
| | | | | | - Tatyana Strekalova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, Russia.,School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht University, Maastricht, The Netherlands.,Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.,Institute of General Pathology and Pathophysiology, University of Würzburg, Moscow, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China.,Laboratory of Petrochemistry, Ural Federal University, Ekaterinburg, Russia
| |
Collapse
|
29
|
Yesudhason BV, Selvan Christyraj JRS, Ganesan M, Subbiahanadar Chelladurai K, Venkatachalam S, Ramalingam A, Benedict J, Paulraj VD, Selvan Christyraj JD. Developmental stages of zebrafish (Danio rerio) embryos and toxicological studies using foldscope microscope. Cell Biol Int 2020; 44:1968-1980. [PMID: 32584484 DOI: 10.1002/cbin.11412] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 01/25/2023]
Abstract
Zebrafish (Danio rerio), is a well-established vertebrate animal model widely used in developmental biology and toxicological research. In the present study, foldscope is used as an innovative tool to study the developmental stages and toxicological analysis of the zebrafish embryos. Briefly, the developmental stages, such as zygote, cleavage, blastula, gastrula, segmentation, and pharyngula formation are observed and documented using simple foldscope. Toxicological parameters upon exposure to different concentration of ethanol extract of Curcuma longa and its lead compound, ar-turmerone along with rhodamine B (bio-coupler) on zebrafish embryos are analyzed upto 72 hr using foldscopes in live condition. The lethal endpoints, such as coagulation, lack of somite formation, non-detachment of tail, and lack of heartbeat are clearly monitored and documented using foldscope. Bio-evaluation of test compounds with the aid of foldscope confirms that the toxicity is directly proportional to the concentration. Our results conclude that, ethanol extract of C. longa, ar-turmerone and rhodamine B exposed embryos remains healthy up to 96, 48, and 24 µg concentrations, respectively. Embryos exposed to higher concentrations become coagulated, however normal physiological active movement of tail lashing and heartbeat are evident in lower concentration exposed embryos. Except coagulation, no other abnormalities are observed and interestingly, the hatching ability is not delayed, when compared with the control embryos. It is confirmed that the test compounds are not highly toxic to zebrafish embryos. Hence it can be used for further analysis, especially for studying the neural-regeneration and its neuronal development in zebrafish embryos.
Collapse
Affiliation(s)
- Beryl Vedha Yesudhason
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| | - Johnson Retnaraj Samuel Selvan Christyraj
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| | - Mijithra Ganesan
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| | - Karthikeyan Subbiahanadar Chelladurai
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| | - Saravanakumar Venkatachalam
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| | - Arun Ramalingam
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| | - Johnson Benedict
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| | - Vennila Devi Paulraj
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| | - Jackson Durairaj Selvan Christyraj
- Regeneration and Stem Cell Biology Lab, Centre for Molecular and Nanomedical Sciences, International Research Centre, Sathyabama Institute of Science and Technology, Chennai, Tamilnadu, India
| |
Collapse
|
30
|
Application of CRISPR Tools for Variant Interpretation and Disease Modeling in Inherited Retinal Dystrophies. Genes (Basel) 2020; 11:genes11050473. [PMID: 32349249 PMCID: PMC7290804 DOI: 10.3390/genes11050473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/27/2022] Open
Abstract
Inherited retinal dystrophies are an assorted group of rare diseases that collectively account for the major cause of visual impairment of genetic origin worldwide. Besides clinically, these vision loss disorders present a high genetic and allelic heterogeneity. To date, over 250 genes have been associated to retinal dystrophies with reported causative variants of every nature (nonsense, missense, frameshift, splice-site, large rearrangements, and so forth). Except for a fistful of mutations, most of them are private and affect one or few families, making it a challenge to ratify the newly identified candidate genes or the pathogenicity of dubious variants in disease-associated loci. A recurrent option involves altering the gene in in vitro or in vivo systems to contrast the resulting phenotype and molecular imprint. To validate specific mutations, the process must rely on simulating the precise genetic change, which, until recently, proved to be a difficult endeavor. The rise of the CRISPR/Cas9 technology and its adaptation for genetic engineering now offers a resourceful suite of tools to alleviate the process of functional studies. Here we review the implementation of these RNA-programmable Cas9 nucleases in culture-based and animal models to elucidate the role of novel genes and variants in retinal dystrophies.
Collapse
|
31
|
Sie ZL, Li RY, Sampurna BP, Hsu PJ, Liu SC, Wang HD, Huang CL, Yuh CH. WNK1 Kinase Stimulates Angiogenesis to Promote Tumor Growth and Metastasis. Cancers (Basel) 2020; 12:cancers12030575. [PMID: 32131390 PMCID: PMC7139507 DOI: 10.3390/cancers12030575] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/23/2022] Open
Abstract
With-no-lysine (K)-1 (WNK1) is the founding member of family of four protein kinases with atypical placement of catalytic lysine that play important roles in regulating epithelial ion transport. Gain-of-function mutations of WNK1 and WNK4 cause a mendelian hypertension and hyperkalemic disease. WNK1 is ubiquitously expressed and essential for embryonic angiogenesis in mice. Increasing evidence indicates the role of WNK kinases in tumorigenesis at least partly by stimulating tumor cell proliferation. Here, we show that human hepatoma cells xenotransplanted into zebrafish produced high levels of vascular endothelial growth factor (VEGF) and WNK1, and induced expression of zebrafish wnk1. Knockdown of wnk1 in zebrafish decreased tumor-induced ectopic vessel formation and inhibited tumor proliferation. Inhibition of WNK1 or its downstream kinases OSR1 (oxidative stress responsive kinase 1)/SPAK (Ste20-related proline alanine rich kinase) using chemical inhibitors decreased ectopic vessel formation as well as proliferation of xenotransplanted hepatoma cells. The effect of WNK and OSR1 inhibitors is greater than that achieved by inhibitor of VEGF signaling cascade. These inhibitors also effectively inhibited tumorigenesis in two separate transgenic zebrafish models of intestinal and hepatocellular carcinomas. Endothelial-specific overexpression of wnk1 enhanced tumorigenesis in transgenic carcinogenic fish, supporting endothelial cell-autonomous effect of WNK1 in tumor promotion. Thus, WNK1 can promote tumorigenesis by multiple effects that include stimulating tumor angiogenesis. Inhibition of WNK1 may be a potent anti-cancer therapy.
Collapse
Affiliation(s)
- Zong-Lin Sie
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (Z.-L.S.); (R.-Y.L.); (B.P.S.); (P.-J.H.)
- Institute of Biotechnology, National Tsing-Hua University, Hsinchu 30013, Taiwan;
| | - Ruei-Yang Li
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (Z.-L.S.); (R.-Y.L.); (B.P.S.); (P.-J.H.)
- Institute of Biotechnology, National Tsing-Hua University, Hsinchu 30013, Taiwan;
| | - Bonifasius Putera Sampurna
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (Z.-L.S.); (R.-Y.L.); (B.P.S.); (P.-J.H.)
| | - Po-Jui Hsu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (Z.-L.S.); (R.-Y.L.); (B.P.S.); (P.-J.H.)
| | - Shu-Chen Liu
- Department of Biomedical Sciences and Engineering, National Central University, Jhongli Dist., Taoyuan 32001, Taiwan;
| | - Horng-Dar Wang
- Institute of Biotechnology, National Tsing-Hua University, Hsinchu 30013, Taiwan;
| | - Chou-Long Huang
- Division of Nephrology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa, IA 52242, USA
- Correspondence: (C.-L.H.); (C.-H.Y.); Tel.: +1-319-356-3972 (C.-L.H.); +011-886-37-206166*35338 (C.-H.Y.)
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli 35053, Taiwan; (Z.-L.S.); (R.-Y.L.); (B.P.S.); (P.-J.H.)
- Institute of Bioinformatics and Structural Biology, National Tsing-Hua University, Hsinchu 30013, Taiwan
- Department of Biological Science & Technology, National Chiao Tung University, Hsinchu 30010, Taiwan
- Ph.D. Program in Environmental and Occupational Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (C.-L.H.); (C.-H.Y.); Tel.: +1-319-356-3972 (C.-L.H.); +011-886-37-206166*35338 (C.-H.Y.)
| |
Collapse
|
32
|
Tonelli F, Bek JW, Besio R, De Clercq A, Leoni L, Salmon P, Coucke PJ, Willaert A, Forlino A. Zebrafish: A Resourceful Vertebrate Model to Investigate Skeletal Disorders. Front Endocrinol (Lausanne) 2020; 11:489. [PMID: 32849280 PMCID: PMC7416647 DOI: 10.3389/fendo.2020.00489] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
Animal models are essential tools for addressing fundamental scientific questions about skeletal diseases and for the development of new therapeutic approaches. Traditionally, mice have been the most common model organism in biomedical research, but their use is hampered by several limitations including complex generation, demanding investigation of early developmental stages, regulatory restrictions on breeding, and high maintenance cost. The zebrafish has been used as an efficient alternative vertebrate model for the study of human skeletal diseases, thanks to its easy genetic manipulation, high fecundity, external fertilization, transparency of rapidly developing embryos, and low maintenance cost. Furthermore, zebrafish share similar skeletal cells and ossification types with mammals. In the last decades, the use of both forward and new reverse genetics techniques has resulted in the generation of many mutant lines carrying skeletal phenotypes associated with human diseases. In addition, transgenic lines expressing fluorescent proteins under bone cell- or pathway- specific promoters enable in vivo imaging of differentiation and signaling at the cellular level. Despite the small size of the zebrafish, many traditional techniques for skeletal phenotyping, such as x-ray and microCT imaging and histological approaches, can be applied using the appropriate equipment and custom protocols. The ability of adult zebrafish to remodel skeletal tissues can be exploited as a unique tool to investigate bone formation and repair. Finally, the permeability of embryos to chemicals dissolved in water, together with the availability of large numbers of small-sized animals makes zebrafish a perfect model for high-throughput bone anabolic drug screening. This review aims to discuss the techniques that make zebrafish a powerful model to investigate the molecular and physiological basis of skeletal disorders.
Collapse
Affiliation(s)
- Francesca Tonelli
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Jan Willem Bek
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Roberta Besio
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Adelbert De Clercq
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Laura Leoni
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Paul J. Coucke
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Andy Willaert
- Department of Biomolecular Medicine, Center of Medical Genetics, Ghent University-University Hospital, Ghent, Belgium
| | - Antonella Forlino
- Biochemistry Unit, Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Antonella Forlino
| |
Collapse
|
33
|
Kang DM, Shin JI, Kim JB, Lee K, Chung JH, Yang HW, Kim KN, Han YS. Detection of 8-oxoguanine and apurinic/apyrimidinic sites using a fluorophore-labeled probe with cell-penetrating ability. BMC Mol Cell Biol 2019; 20:54. [PMID: 31775627 PMCID: PMC6881995 DOI: 10.1186/s12860-019-0236-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/14/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Reactive oxygen species (ROS) produce different lesions in DNA by ROS-induced DNA damage. Detection and quantification of 8-oxo-7,8-dihydroguanine (8-oxoG) within cells are important for study. Human ribosomal protein S3 (hRpS3) has a high binding affinity to 8-oxoG. In this study, we developed an imaging probe to detect 8-oxoG using a specific peptide from hRpS3. Transactivator (TAT) proteins are known to have cell-penetrating properties. Therefore, we developed a TAT-S3 probe by attaching a TAT peptide to our imaging probe. RESULTS A DNA binding assay was conducted to confirm that our probe bound to 8-oxoG and apurinic/apyrimidinic (AP) sites. We confirmed that the TAT-S3 probe localized in the mitochondria, without permeabilization, and fluoresced in H2O2-treated HeLa cells and zebrafish embryos. Treatment with Mitoquinone (MitoQ), a mitochondria-targeted antioxidant, reduced TAT-S3 probe fluorescence. Additionally, treatment with O8, an inhibitor of OGG1, increased probe fluorescence. A competition assay was conducted with an aldehyde reaction probe (ARP) and methoxyamine (MX) to confirm binding of TAT-S3 to the AP sites. The TAT-S3 probe showed competitive binding to AP sites with ARP and MX. CONCLUSIONS These results revealed that the TAT-S3 probe successfully detected the presence of 8-oxoG and AP sites in damaged cells. The TAT-S3 probe may have applications for the detection of diseases caused by reactive oxygen species.
Collapse
Affiliation(s)
- Dong Min Kang
- Department of Advanced Technology Fusion, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Jong-Il Shin
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Ji Beom Kim
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Kyungho Lee
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Ji Hyung Chung
- Department of Applied Bioscience, College of Life Science, CHA University, Pocheon, 11160, South Korea
| | - Hye-Won Yang
- Department of Marine Life Science, Jeju National University, Jeju, 63243, South Korea
| | - Kil-Nam Kim
- Chuncheon Center, Korea Basic Science Institute (KBSI), Chuncheon, 24341, South Korea
| | - Ye Sun Han
- Department of Advanced Technology Fusion, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
| |
Collapse
|
34
|
Insensitivity to reward shifts in zebrafish (Danio rerio) and implications for assessing affective states. Anim Cogn 2019; 23:87-100. [PMID: 31722040 DOI: 10.1007/s10071-019-01318-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 06/06/2019] [Accepted: 10/09/2019] [Indexed: 10/25/2022]
Abstract
Theory and empirical findings predict that individuals in a negative affective state are more sensitive to unexpected reward loss and less sensitive to unexpected reward gain compared to individuals in a neutral or positive affective state. We explore the use of sensitivity to reward shifts measured during successive contrast tasks as an indicator of affect in zebrafish (Danio rerio). In line with the assumption that exposure to rewarding stimuli induces a relatively positive affective state compared to exposure to stimuli that they do not prefer, we confirmed that zebrafish prefer enriched over barren environments, suggesting that the enriched environment is associated with positive affective states. We trained individuals to swim down a channel for food rewards of differing value and then presented them with unexpected increases or decreases in reward value. Contrary to our hypothesis, individuals conditioned to a high-value reward continued swimming at the same speed when reward value was downshifted, thus showing no successive negative contrast effect and appearing insensitive to reward loss. Individuals whose rewards were upshifted gradually increased their speed, but did not display successive positive contrast effects typical of sensitivity to reward gains. In both cases, housing type did not result in differences in swim time. One potential explanation is that goal-directed control of behaviour is necessary for an animal to show a successive contrast response to unexpected reward gain or loss, and the behaviour of zebrafish in this task was under habitual control, perhaps due to over-training. If so, refinements to task design and training procedures will allow further progress with this assay.
Collapse
|
35
|
Sieber S, Grossen P, Bussmann J, Campbell F, Kros A, Witzigmann D, Huwyler J. Zebrafish as a preclinical in vivo screening model for nanomedicines. Adv Drug Deliv Rev 2019; 151-152:152-168. [PMID: 30615917 DOI: 10.1016/j.addr.2019.01.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/23/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022]
Abstract
The interactions of nanomedicines with biological environments is heavily influenced by their physicochemical properties. Formulation design and optimization are therefore key steps towards successful nanomedicine development. Unfortunately, detailed assessment of nanomedicine formulations, at a macromolecular level, in rodents is severely limited by the restricted imaging possibilities within these animals. Moreover, rodent in vivo studies are time consuming and expensive, limiting the number of formulations that can be practically assessed in any one study. Consequently, screening and optimisation of nanomedicine formulations is most commonly performed in surrogate biological model systems, such as human-derived cell cultures. However, despite the time and cost advantages of classical in vitro models, these artificial systems fail to reflect and mimic the complex biological situation a nanomedicine will encounter in vivo. This has acutely hampered the selection of potentially successful nanomedicines for subsequent rodent in vivo studies. Recently, zebrafish have emerged as a promising in vivo model, within nanomedicine development pipelines, by offering opportunities to quickly screen nanomedicines under in vivo conditions and in a cost-effective manner so as to bridge the current gap between in vitro and rodent studies. In this review, we outline several advantageous features of the zebrafish model, such as biological conservation, imaging modalities, availability of genetic tools and disease models, as well as their various applications in nanomedicine development. Critical experimental parameters are discussed and the most beneficial applications of the zebrafish model, in the context of nanomedicine development, are highlighted.
Collapse
Affiliation(s)
- Sandro Sieber
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland
| | - Jeroen Bussmann
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Frederick Campbell
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Alexander Kros
- Department of Supramolecular and Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland; Department of Biochemistry and Molecular Biology, University of British Columbia, Health Sciences Mall, Vancouver, British Columbia, Canada..
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
36
|
Wang R, Zhang H, Du J, Xu J. Heat resilience in embryonic zebrafish revealed using an in vivo stress granule reporter. J Cell Sci 2019; 132:jcs.234807. [PMID: 31558681 PMCID: PMC6826007 DOI: 10.1242/jcs.234807] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/19/2019] [Indexed: 12/13/2022] Open
Abstract
Although the regulation of stress granules has become an intensely studied topic, current investigations of stress granule assembly, disassembly and dynamics are mainly performed in cultured cells. Here, we report the establishment of a stress granule reporter to facilitate the real-time study of stress granules in vivo. Using CRISPR/Cas9, we fused a green fluorescence protein (GFP) to endogenous G3BP1 in zebrafish. The GFP–G3BP1 reporter faithfully and robustly responded to heat stress in zebrafish embryos and larvae. The induction of stress granules varied by brain regions under the same stress condition, with the midbrain cells showing the highest efficiency and dynamics. Furthermore, pre-conditioning using lower heat stress significantly limited stress granule formation during subsequent higher heat stress. More interestingly, stress granule formation was much more robust in zebrafish embryos than in larvae and coincided with significantly elevated levels of phosphorylated eIF2α and enhanced heat resilience. Therefore, these findings have generated new insights into stress response in zebrafish during early development and demonstrated that the GFP–G3BP1 knock-in zebrafish could be a valuable tool for the investigation of stress granule biology. This article has an associated First Person interview with the first author of the paper. Summary: Establishment of a new transgenic zebrafish line with knock-in GFP-G3BP1 to visualize stress granule dynamics in live animals in real time.
Collapse
Affiliation(s)
- Ruiqi Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Hefei Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai 200031, China
| | - Jin Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| |
Collapse
|
37
|
Kaitetzidou E, Katsiadaki I, Lagnel J, Antonopoulou E, Sarropoulou E. Unravelling paralogous gene expression dynamics during three-spined stickleback embryogenesis. Sci Rep 2019; 9:3752. [PMID: 30842559 PMCID: PMC6403355 DOI: 10.1038/s41598-019-40127-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/08/2019] [Indexed: 12/24/2022] Open
Abstract
Development requires the implementation of a plethora of molecular mechanisms, involving a large set of genes to ensure proper cell differentiation, morphogenesis of tissues and organs as well as the growth of the organism. Genome duplication and resulting paralogs are considered to provide the raw genetic materials important for new adaptation opportunities and boosting evolutionary innovation. The present study investigated paralogous genes, involved in three-spined stickleback (Gasterosteus aculeatus) development. Therefore, the transcriptomes of five early stages comprising developmental leaps were explored. Obtained expression profiles reflected the embryo's needs at different stages. Early stages, such as the morula stage comprised transcripts mainly involved in energy requirements while later stages were mostly associated with GO terms relevant to organ development and morphogenesis. The generated transcriptome profiles were further explored for differential expression of known and new paralogous genes. Special attention was given to hox genes, with hoxa13a being of particular interest and to pigmentation genes where itgb1, involved in the melanophore development, displayed a complementary expression pattern throughout studied stages. Knowledge obtained by untangling specific paralogous gene functions during development might not only significantly contribute to the understanding of teleost ontogenesis but might also shed light on paralogous gene evolution.
Collapse
Affiliation(s)
- Elisavet Kaitetzidou
- Department of Zoology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Institute for Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece
| | - Ioanna Katsiadaki
- Centre for Environment Fisheries and Aquaculture Science, (Cefas), Weymouth, UK
| | - Jacques Lagnel
- Institute for Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece.,Institut National de la Recherche Agronomique (INRA), Génétique et Amélioration des Fruits et Légumes (GALF), Montfavet Cedex, France
| | - Efthimia Antonopoulou
- Department of Zoology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Elena Sarropoulou
- Institute for Marine Biology, Biotechnology and Aquaculture (IMBBC), Hellenic Centre for Marine Research (HCMR), Heraklion, Greece.
| |
Collapse
|
38
|
Li JJ, Zhang Y, Han LW, Tian QP, He QX, Wang XM, Sun C, Han J, Liu KC. Tenacissoside H exerts an anti-inflammatory effect by regulating the nf-κb and p38 pathways in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2018; 83:205-212. [PMID: 30213642 DOI: 10.1016/j.fsi.2018.09.032] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 08/26/2018] [Accepted: 09/10/2018] [Indexed: 06/08/2023]
Abstract
Marsdenia tenacissima exhibits biological activity with heat-clearing and detoxifying properties, relieving coughs and asthma and exerting anticancer and anti-HIV effects. Tenacissioside H (TH) is a Chinese medicine monomer extracted from the dried stem of Marsdenia tenacissima. We investigated the in vivo anti-inflammatory activity of TH using three different zebrafish inflammation models: local inflammation induced by tail cutting, acute inflammation induced by CuSO4, and systemic inflammation induced by lipopolysaccharide (LPS). Real time-polymerase chain reaction (RT-PCR) was used to elucidate the mechanism of TH action against LPS-induced inflammatory responses. Our results showed TH significantly reduced the number of macrophages in the injured zebrafish tail, inhibited CuSO4-induced migration of macrophages toward the neural mound, and decreased the distribution of macrophages in tail fin compared to LPS-treated group. Furthermore, TH inhibits LPS-induced inflammation responses in zebrafish by modulating the nuclear factor κB (nf-κb) and p38 pathways to regulate inflammatory cytokines, such as tumor necrosis factor-α (tnf-α), cyclooxygenase (cox-2), interleukin-1b (il-1b), interleukin-8 (il-8), interleukin-10 (il-10), nitric oxide synthase (nos2b) and prostaglandin E synthase (ptges). In conclusion, TH possesses anti-inflammation activity via the regulation of the nf-κb and p38 pathways. This finding provides a reference for the clinical application of Xiaoaiping (the trade name of Marsdenia tenacissima extract).
Collapse
Affiliation(s)
- Juan-Juan Li
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China; Shanxi Medical University, 56 Xinjiannan Road, Yingze District, Taiyuan, 030001, Shanxi Province, PR China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Key Laboaratory for Biosensor of Shandong Province, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| | - Li-Wen Han
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Key Laboaratory for Biosensor of Shandong Province, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Qing-Ping Tian
- Shanxi Medical University, 56 Xinjiannan Road, Yingze District, Taiyuan, 030001, Shanxi Province, PR China
| | - Qiu-Xia He
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Key Laboaratory for Biosensor of Shandong Province, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Xi-Min Wang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Key Laboaratory for Biosensor of Shandong Province, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Chen Sun
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Key Laboaratory for Biosensor of Shandong Province, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Jian Han
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Key Laboaratory for Biosensor of Shandong Province, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China
| | - Ke-Chun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China; Key Laboratory for Drug Screening Technology of Shandong Academy of Sciences, Key Laboaratory for Biosensor of Shandong Province, Qilu University of Technology (Shandong Academy of Sciences), 28789 Jingshidong Road, Licheng District, Jinan 250103, Shandong Province, PR China.
| |
Collapse
|
39
|
Insights into Endothelial Progenitor Cells: Origin, Classification, Potentials, and Prospects. Stem Cells Int 2018; 2018:9847015. [PMID: 30581475 PMCID: PMC6276490 DOI: 10.1155/2018/9847015] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/27/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023] Open
Abstract
With the discovery of endothelial progenitor cells (EPCs) in the late 1990s, a paradigm shift in the concept of neoangiogenesis occurred. The identification of circulating EPCs in peripheral blood marked the beginning of a new era with enormous potential in the rapidly transforming regenerative field. Overwhelmed with the revelation, researchers across the globe focused on isolating, defining, and interpreting the role of EPCs in various physiological and pathological conditions. Consequently, controversies emerged regarding the isolation techniques and classification of EPCs. Nevertheless, the potential of using EPCs in tissue engineering as an angiogenic source has been extensively explored. Concomitantly, the impact of EPCs on various diseases, such as diabetes, cancer, and cardiovascular diseases, has been studied. Within the limitations of the current knowledge, this review attempts to delineate the concept of EPCs in a sequential manner from the speculative history to a definitive presence (origin, sources of EPCs, isolation, and identification) and significance of these EPCs. Additionally, this review is aimed at serving as a guide for investigators, identifying potential research gaps, and summarizing our current and future prospects regarding EPCs.
Collapse
|
40
|
Zebrafish: an emerging real-time model system to study Alzheimer's disease and neurospecific drug discovery. Cell Death Discov 2018; 4:45. [PMID: 30302279 PMCID: PMC6170431 DOI: 10.1038/s41420-018-0109-7] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/21/2018] [Accepted: 08/23/2018] [Indexed: 12/22/2022] Open
Abstract
Zebrafish (Danio rerio) is emerging as an increasingly successful model for translational research on human neurological disorders. In this review, we appraise the high degree of neurological and behavioural resemblance of zebrafish with humans. It is highly validated as a powerful vertebrate model for investigating human neurodegenerative diseases. The neuroanatomic and neurochemical pathways of zebrafish brain exhibit a profound resemblance with the human brain. Physiological, emotional and social behavioural pattern similarities between them have also been well established. Interestingly, zebrafish models have been used successfully to simulate the pathology of Alzheimer’s disease (AD) as well as Tauopathy. Their relatively simple nervous system and the optical transparency of the embryos permit real-time neurological imaging. Here, we further elaborate on the use of recent real-time imaging techniques to obtain vital insights into the neurodegeneration that occurs in AD. Zebrafish is adeptly suitable for Ca2+ imaging, which provides a better understanding of neuronal activity and axonal dystrophy in a non-invasive manner. Three-dimensional imaging in zebrafish is a rapidly evolving technique, which allows the visualisation of the whole organism for an elaborate in vivo functional and neurophysiological analysis in disease condition. Suitability to high-throughput screening and similarity with humans makes zebrafish an excellent model for screening neurospecific compounds. Thus, the zebrafish model can be pivotal in bridging the gap from the bench to the bedside. This fish is becoming an increasingly successful model to understand AD with further scope for investigation in neurodevelopment and neurodegeneration, which promises exciting research opportunities in the future.
Collapse
|
41
|
Lee W, Kim EA, Um JH, Kang N, Oh JY, Park SY, Han EJ, Cheong SH, Chang KJ, Kim SH, Ahn CB, Jeon YJ, Ahn G. Protective Effects of Xylose-Taurine Reduced against Damages Caused by Oxidative Stress in Zebrafish Embryos In Vivo Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:643-653. [PMID: 28849488 DOI: 10.1007/978-94-024-1079-2_50] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
The zebrafish (Danio rerio) is useful and convenient vertebrate models in various studies in human disease and drug discovery. In this present study, we first evaluated whether Xylose-Taurine reduced (X-T-R), a taurine derivate protects zebrafish embryos against oxidative stress caused by AAPH (2,2'-Azobis(2-amidinopropane) dihydrochloride). First of all, we selected the concentration of X-T-R showing no toxicity in zebrafish embryos. We identified that X-T-R significantly increased the survival of zebrafish embryo reduced by treatment of AAPH. Also, X-T-R effectively inhibited the productions of reactive oxygen species (ROS) and nitric oxide (NO) as well as the formation of cell death in zebrafish embryos. Moreover, X-T-R down-regulated the expression levels of Bax, caspase-3, caspase-9 and p53 known as pro-apoptotic molecules, whereas up-regulated those of Bcl-2, an anti-apoptotic molecule in AAPH-treated zebrafish embryos. From these results, this study reveals that X-T-R, a taurine derivate might be a potential protector against various damages caused by oxidative stress.
Collapse
Affiliation(s)
- WonWoo Lee
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Eun-A Kim
- Jeju International Marine Science Center for Research and Education, Korea Institute of Ocean Science & Technology (KIOST), Jeju, Republic of Korea
| | - Ju Hyung Um
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu, Republic of Korea
| | - Nalae Kang
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu, Republic of Korea
| | - Jae Young Oh
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea
| | - Soo Yeon Park
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu, Republic of Korea
| | - Eui Jeong Han
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu, Republic of Korea
| | - Sun Hee Cheong
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu, Republic of Korea
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu, Republic of Korea
| | - Kyung Ja Chang
- Department of Food and Nutrition, Inha University, Incheon, Republic of Korea
| | - Sung Hoon Kim
- Department of Chemistry, Konkuk University, Seoul, Republic of Korea
| | - Chang-Bum Ahn
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju, Republic of Korea.
| | - Ginnae Ahn
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu, Republic of Korea.
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu, Republic of Korea.
| |
Collapse
|
42
|
Wojtczyk-Miaskowska A, Schlichtholz B. DNA damage and oxidative stress in long-lived aquatic organisms. DNA Repair (Amst) 2018; 69:14-23. [DOI: 10.1016/j.dnarep.2018.07.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/09/2018] [Indexed: 12/11/2022]
|
43
|
Cornet C, Di Donato V, Terriente J. Combining Zebrafish and CRISPR/Cas9: Toward a More Efficient Drug Discovery Pipeline. Front Pharmacol 2018; 9:703. [PMID: 30018554 PMCID: PMC6037853 DOI: 10.3389/fphar.2018.00703] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/11/2018] [Indexed: 12/13/2022] Open
Abstract
The use of zebrafish larvae in basic and applied research has grown exponentially during the last 20 years. The reasons for this success lay in its specific experimental advantages: on the one hand, the small size, the large number of progeny and the fast life cycle greatly facilitate large-scale approaches while maintaining 3Rs amenability; on the other hand, high genetic and physiological homology with humans and ease of genetic manipulation make zebrafish larvae a highly robust model for understanding human disease. Together, these advantages allow using zebrafish larvae for performing high-throughput research, both in terms of chemical and genetic phenotypic screenings. Therefore, the zebrafish larva as an animal model is placed between more reductionist in vitro high-throughput screenings and informative but low-throughput preclinical assays using mammals. However, despite its biological advantages and growing translational validation, zebrafish remains scarcely used in current drug discovery pipelines. In a context in which the pharmaceutical industry is facing a productivity crisis in bringing new drugs to the market, the combined advantages of zebrafish and the CRISPR/Cas9 system, the most powerful technology for genomic editing to date, has the potential to become a valuable tool for streamlining the generation of models mimicking human disease, the validation of novel drug targets and the discovery of new therapeutics. This review will focus on the most recent advances on CRISPR/Cas9 implementation in zebrafish and all their potential uses in biomedical research and drug discovery.
Collapse
Affiliation(s)
- Carles Cornet
- ZeClinics SL, PRBB (Barcelona Biomedical Research Park), Barcelona, Spain
| | - Vincenzo Di Donato
- ZeClinics SL, PRBB (Barcelona Biomedical Research Park), Barcelona, Spain
| | - Javier Terriente
- ZeClinics SL, PRBB (Barcelona Biomedical Research Park), Barcelona, Spain
| |
Collapse
|
44
|
Compound K Inhibits the Lipopolysaccharide-Induced Inflammatory Responses in Raw 264.7 Cell Line and Zebrafish. APPLIED SCIENCES-BASEL 2018. [DOI: 10.3390/app8060924] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
45
|
Kang CP, Tu HC, Fu TF, Wu JM, Chu PH, Chang DTH. An automatic method to calculate heart rate from zebrafish larval cardiac videos. BMC Bioinformatics 2018; 19:169. [PMID: 29743010 PMCID: PMC5944013 DOI: 10.1186/s12859-018-2166-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/25/2018] [Indexed: 11/29/2022] Open
Abstract
Background Zebrafish is a widely used model organism for studying heart development and cardiac-related pathogenesis. With the ability of surviving without a functional circulation at larval stages, strong genetic similarity between zebrafish and mammals, prolific reproduction and optically transparent embryos, zebrafish is powerful in modeling mammalian cardiac physiology and pathology as well as in large-scale high throughput screening. However, an economical and convenient tool for rapid evaluation of fish cardiac function is still in need. There have been several image analysis methods to assess cardiac functions in zebrafish embryos/larvae, but they are still improvable to reduce manual intervention in the entire process. This work developed a fully automatic method to calculate heart rate, an important parameter to analyze cardiac function, from videos. It contains several filters to identify the heart region, to reduce video noise and to calculate heart rates. Results The proposed method was evaluated with 32 zebrafish larval cardiac videos that were recording at three-day post-fertilization. The heart rate measured by the proposed method was comparable to that determined by manual counting. The experimental results show that the proposed method does not lose accuracy while largely reducing the labor cost and uncertainty of manual counting. Conclusions With the proposed method, researchers do not have to manually select a region of interest before analyzing videos. Moreover, filters designed to reduce video noise can alleviate background fluctuations during the video recording stage (e.g. shifting), which makes recorders generate usable videos easily and therefore reduce manual efforts while recording. Electronic supplementary material The online version of this article (10.1186/s12859-018-2166-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chia-Pin Kang
- Department of Electronic Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Hung-Chi Tu
- The Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Tzu-Fun Fu
- The Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Jhe-Ming Wu
- Department of Electronic Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Po-Hsun Chu
- Department of Electronic Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Darby Tien-Hao Chang
- Department of Electronic Engineering, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
46
|
Affiliation(s)
- Jason R. Meyers
- Department of Biology and Program in Neuroscience, Colgate University; Hamilton New York
| |
Collapse
|
47
|
Lobos-Ruiz P, Araya G, Monasterio O, Pouchucq L. Induction of protein aggregation in zebrafish embryos as a method for the screening of new drugs or mutations against proteinopathies. MethodsX 2018; 5:322-327. [PMID: 30046518 PMCID: PMC6058008 DOI: 10.1016/j.mex.2018.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 04/04/2018] [Indexed: 11/19/2022] Open
Abstract
The sustained increase in the prevalence of protein aggregation related diseases requires the development of feasible methods for the design of therapeutic alternatives. The procedure traditionally used for the search of drugs or therapeutic mutations includes in vitro experiments, designed to prevent the aggregation of model proteins, which are then complemented with cellular toxicity studies in vivo, slowing down the finding of solutions. To address this, we have developed a protocol that facilitates the search of molecules and anti-aggregation mutations since it allows to evaluate their therapeutic capabilities directly in in vivo experiments with the use of zebrafish early embryos. Avoiding the necessity of performing in vitro and in vivo procedures separately. Giving a more realistic method for the results interpretation. Zebrafish embryos were induced to produce intracellular aggregates of proteins by simple microinjections of known quantities of an aggregation prone protein previously labeled. The toxicity was evaluated by the survival of the embryos, while the formation of aggregates was quantified by fluorescence microscopy. The size distribution of the protein aggregates was revealed by means of ultracentrifuge sedimentation analysis. For the development of the present method, the human γ-tubulin protein was used as model protein, which generated intracellular aggregates in more than 60% of the injected embryos. To evaluate the method, a mutation was performed that altered the state of intracellular aggregation of γ-tubulin, obtaining a significant decrease in the amount and size of the intracellular aggregates. The present method can be used for any suitable intracellular aggregation protein model. The current method present important advantages such as: Easy induction of intracellular aggregates. Simple detection of intracellular protein aggregates through fluorescence microscopy and subcellular fractionation. Overall view of the effect of drugs or mutations by combining the toxicity, the development behavior and the size distribution of intracellular protein aggregates.
Collapse
Affiliation(s)
- Pablo Lobos-Ruiz
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gissela Araya
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Octavio Monasterio
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Luis Pouchucq
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
- Laboratorio de Biotecnología Vegetal Ambiental, Departamento de Biotecnología, Universidad Tecnológica Metropolitana, Santiago, Chile
- Corresponding author at: Las Palmeras, 3360, Ñuñoa, Santiago, Chile.
| |
Collapse
|
48
|
Schüttler A, Reiche K, Altenburger R, Busch W. The Transcriptome of the Zebrafish Embryo After Chemical Exposure: A Meta-Analysis. Toxicol Sci 2018; 157:291-304. [PMID: 28329862 PMCID: PMC5443304 DOI: 10.1093/toxsci/kfx045] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Numerous studies have been published in the past years investigating the transcriptome of the zebrafish embryo (ZFE) upon being subjected to chemical stress. Aiming at a more mechanistic understanding of the results of such studies, knowledge about commonalities of transcript regulation in response to chemical stress is needed. Thus, our goal in this study was to identify and interpret genes and gene sets constituting a general response to chemical exposure. Therefore, we aggregated and reanalyzed published toxicogenomics data obtained with the ZFE. We found that overlap of differentially transcribed genes in response to chemical stress across independent studies is generally low and the most commonly differentially transcribed genes appear in less than 50% of all treatments across studies. However, effect size analysis revealed several genes showing a common trend of differential expression, among which genes related to calcium homeostasis emerged as key, especially in exposure settings up to 24 h post-fertilization. Additionally, we found that these and other downregulated genes are often linked to anatomical regions developing during the respective exposure period. Genes showing a trend of increased expression were, among others, linked to signaling pathways (e.g., Wnt, Fgf) as well as lysosomal structures and apoptosis. The findings of this study help to increase the understanding of chemical stress responses in the developing zebrafish embryo and provide a starting point to improve experimental designs for this model system. In future, improved time- and concentration-resolved experiments should offer better understanding of stress response patterns and access to mechanistic information.
Collapse
Affiliation(s)
- Andreas Schüttler
- Department Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research - UFZ, Permoserstraβe 15, Leipig, Germany.,Institute for Environmental Research, RWTH Aachen, Worringerweg 1, Aachen, Germany
| | - Kristin Reiche
- Young Investigators Group Bioinformatics and Transcriptomics, Department Molecular Systems Biology, Helmholtz Centre for Environmental Research - UFZ, Permoserstraβe 15, Leipig, Germany.,Bioinformatics Unit, Department of Diagnostics, Fraunhofer Institute for Cell Therapy and Immunology, Perlickstraβe 1, Leipzig, Germany
| | - Rolf Altenburger
- Department Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research - UFZ, Permoserstraβe 15, Leipig, Germany.,Institute for Environmental Research, RWTH Aachen, Worringerweg 1, Aachen, Germany
| | - Wibke Busch
- Department Bioanalytical Ecotoxicology, Helmholtz Centre for Environmental Research - UFZ, Permoserstraße 15, Leipig, Germany
| |
Collapse
|
49
|
Pouchucq L, Lobos-Ruiz P, Araya G, Valpuesta JM, Monasterio O. The chaperonin CCT promotes the formation of fibrillar aggregates of γ-tubulin. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2018; 1866:519-526. [PMID: 29339327 DOI: 10.1016/j.bbapap.2018.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 12/29/2022]
Abstract
The type II chaperonin CCT is involved in the prevention of the pathogenesis of numerous human misfolding disorders, as it sequesters misfolded proteins, blocks their aggregation and helps them to achieve their native state. In addition, it has been reported that CCT can prevent the toxicity of non-client amyloidogenic proteins by the induction of non-toxic aggregates, leading to new insight in chaperonin function as an aggregate remodeling factor. Here we add experimental evidence to this alternative mechanism by which CCT actively promotes the formation of conformationally different aggregates of γ-tubulin, a non-amyloidogenic CCT client protein, which are mediated by specific CCT-γ-tubulin interactions. The in vitro-induced aggregates were in some cases long fiber polymers, which compete with the amorphous aggregates. Direct injection of unfolded purified γ-tubulin into single-cell zebra fish embryos allowed us to relate this in vitro activity with the in vivo formation of intracellular aggregates. Injection of a CCT-binding deficient γ-tubulin mutant dramatically diminished the size of the intracellular aggregates, increasing the toxicity of the misfolded protein. These results point to CCT having a role in the remodeling of aggregates, constituting one of its many functions in cellular proteostasis.
Collapse
Affiliation(s)
- Luis Pouchucq
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile; Laboratorio de Biotecnología Vegetal Ambiental, Departamento de Biotecnología, Universidad Tecnológica Metropolitana, Santiago, Chile
| | - Pablo Lobos-Ruiz
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Gissela Araya
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - José María Valpuesta
- Departamento de Estructura de Macromoléculas, Centro Nacional de Biotecnología (CNB-CSIC), Campus de la Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Octavio Monasterio
- Laboratorio de Biología Estructural y Molecular, Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
50
|
Basta HA, Buzak AJ, McClure MA. Identification of Novel Retroid Agents in Danio rerio, Oryzias latipes, Gasterosteus aculeatus and Tetraodon nigroviridis. Evol Bioinform Online 2017. [DOI: 10.1177/117693430700300018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Retroid agents are genomes that encode a reverse transcriptase (RT) and replicate or transpose by way of an RNA intermediate. The Genome Parsing Suite (GPS) is software created to identify and characterize Retroid agents in any genome database (McClure et al. 2005). The detailed analysis of all Retroid agents found by the GPS in Danio rerio (zebrafish), Oryzias latipes (medaka), Gasterosteus aculeatus (stickleback) and Tetraodon nigroviridis (spotted green pufferfish) reveals extensive Retroid agent diversity in the compact genomes of all four fish. Novel Retroid agents were identified by the GPS software: the telomerase reverse transcriptase (TERT) in O. latipes, G aculeatus and T. nigroviridis and a potential TERT in D. rerio, a retrotransposon in D. rerio, and multiple lineages of endogenous retroviruses (ERVs) in D. rerio, O. latipes and G aculeatus.
Collapse
Affiliation(s)
- Holly A. Basta
- Department of Microbiology and the Center for Computational Biology, Montana State University at Bozeman, 109 Lewis Hall, Bozeman, MT 59717, U.S.A
| | - Alex J. Buzak
- Department of Microbiology and the Center for Computational Biology, Montana State University at Bozeman, 109 Lewis Hall, Bozeman, MT 59717, U.S.A
| | - Marcella A. McClure
- Department of Microbiology and the Center for Computational Biology, Montana State University at Bozeman, 109 Lewis Hall, Bozeman, MT 59717, U.S.A
| |
Collapse
|