1
|
Deng X, Chen Y, Duan Q, Ding J, Wang Z, Wang J, Chen X, Zhou L, Zhao L. Genetic and molecular mechanisms of hydrocephalus. Front Mol Neurosci 2025; 17:1512455. [PMID: 39839745 PMCID: PMC11746911 DOI: 10.3389/fnmol.2024.1512455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 12/20/2024] [Indexed: 01/23/2025] Open
Abstract
Hydrocephalus is a neurological condition caused by aberrant circulation and/or obstructed cerebrospinal fluid (CSF) flow after cerebral ventricle abnormal dilatation. In the past 50 years, the diagnosis and treatment of hydrocephalus have remained understudied and underreported, and little progress has been made with respect to prevention or treatment. Further research on the pathogenesis of hydrocephalus is essential for developing new diagnostic, preventive, and therapeutic strategies. Various genetic and molecular abnormalities contribute to the mechanisms of hydrocephalus, including gene deletions or mutations, the activation of cellular inflammatory signaling pathways, alterations in water channel proteins, and disruptions in iron metabolism. Several studies have demonstrated that modulating the expression of key proteins, including TGF-β, VEGF, Wnt, AQP, NF-κB, and NKCC, can significantly influence the onset and progression of hydrocephalus. This review summarizes and discusses key mechanisms that may be involved in the pathogenesis of hydrocephalus at both the genetic and molecular levels. While obstructive hydrocephalus can often be addressed by removing the obstruction, most cases require treatment strategies that involve merely slowing disease progression by correcting CSF circulation patterns. There have been few new research breakthroughs in the prevention and treatment of hydrocephalus.
Collapse
Affiliation(s)
- Xuehai Deng
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Yiqian Chen
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Qiyue Duan
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Jianlin Ding
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Zhong Wang
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- School of Clinical Medicine, North Sichuan Medical College, Nanchong, China
| | - Junchi Wang
- School of Dentistry, North Sichuan Medical College, Nanchong, China
| | - Xinlong Chen
- Department of Neurosurgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Long Zhao
- Department of Neurosurgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
2
|
Wang K, Tang Z, Yang Y, Guo Y, Liu Z, Su Z, Li X, Xiao G. Zebrafish as a Model Organism for Congenital Hydrocephalus: Characteristics and Insights. Zebrafish 2024; 21:361-384. [PMID: 39510565 DOI: 10.1089/zeb.2024.0148] [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: 11/15/2024] Open
Abstract
Hydrocephalus is a cerebrospinal fluid-related disease that usually manifests as abnormal dilation of the ventricles, with a triad of clinical findings including walking difficulty, reduced attention span, and urinary frequency or incontinence. The onset of congenital hydrocephalus is closely related to mutations in genes that regulate brain development. Currently, our understanding of the mechanisms of congenital hydrocephalus remains limited, and the prognosis of existing treatments is unsatisfactory. Additionally, there are no suitable or dedicated model organisms for congenital hydrocephalus. Therefore, it is significant to determine the mechanism and develop special animal models of congenital hydrocephalus. Recently, zebrafish have emerged as a popular model organism in many fields, including developmental biology, genetics, and toxicology. Its genome shares high similarity with that of humans, and it has fast and low-cost reproduction. These advantages make it suitable for studying the pathogenesis and therapeutic approaches for various diseases, specifically congenital diseases. This study explored the possibility of using zebrafish as a model organism for congenital hydrocephalus. This review describes the characteristics of zebrafish and discusses specific congenital hydrocephalus models. The advantages and limitations of using zebrafish for hydrocephalus research are highlighted, and insights for further model development are provided.
Collapse
Affiliation(s)
- Kaiyue Wang
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhi Tang
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
| | - Yijian Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Yating Guo
- Department of Neurosurgery, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Zhangjie Su
- Department of Neurosurgery, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, United Kingdom
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| | - Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, PR China
- Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, PR China
| |
Collapse
|
3
|
Liu XY, Song X, Czosnyka M, Robba C, Czosnyka Z, Summers JL, Yu HJ, Gao GY, Smielewski P, Guo F, Pang MJ, Ming D. Congenital hydrocephalus: a review of recent advances in genetic etiology and molecular mechanisms. Mil Med Res 2024; 11:54. [PMID: 39135208 PMCID: PMC11318184 DOI: 10.1186/s40779-024-00560-5] [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: 03/09/2024] [Accepted: 07/28/2024] [Indexed: 08/15/2024] Open
Abstract
The global prevalence rate for congenital hydrocephalus (CH) is approximately one out of every five hundred births with multifaceted predisposing factors at play. Genetic influences stand as a major contributor to CH pathogenesis, and epidemiological evidence suggests their involvement in up to 40% of all cases observed globally. Knowledge about an individual's genetic susceptibility can significantly improve prognostic precision while aiding clinical decision-making processes. However, the precise genetic etiology has only been pinpointed in fewer than 5% of human instances. More occurrences of CH cases are required for comprehensive gene sequencing aimed at uncovering additional potential genetic loci. A deeper comprehension of its underlying genetics may offer invaluable insights into the molecular and cellular basis of this brain disorder. This review provides a summary of pertinent genes identified through gene sequencing technologies in humans, in addition to the 4 genes currently associated with CH (two X-linked genes L1CAM and AP1S2, two autosomal recessive MPDZ and CCDC88C). Others predominantly participate in aqueduct abnormalities, ciliary movement, and nervous system development. The prospective CH-related genes revealed through animal model gene-editing techniques are further outlined, focusing mainly on 4 pathways, namely cilia synthesis and movement, ion channels and transportation, Reissner's fiber (RF) synthesis, cell apoptosis, and neurogenesis. Notably, the proper functioning of motile cilia provides significant impulsion for cerebrospinal fluid (CSF) circulation within the brain ventricles while mutations in cilia-related genes constitute a primary cause underlying this condition. So far, only a limited number of CH-associated genes have been identified in humans. The integration of genotype and phenotype for disease diagnosis represents a new trend in the medical field. Animal models provide insights into the pathogenesis of CH and contribute to our understanding of its association with related complications, such as renal cysts, scoliosis, and cardiomyopathy, as these genes may also play a role in the development of these diseases. Genes discovered in animals present potential targets for new treatments but require further validation through future human studies.
Collapse
Affiliation(s)
- Xiu-Yun Liu
- Medical School, Tianjin University, Tianjin, 300072, China
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, 300072, China
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300380, China
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Xin Song
- Medical School, Tianjin University, Tianjin, 300072, China
| | - Marek Czosnyka
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Chiara Robba
- San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, 16132, Genoa, Italy
| | - Zofia Czosnyka
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Jennifer Lee Summers
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Hui-Jie Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Guo-Yi Gao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Peter Smielewski
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Fang Guo
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Mei-Jun Pang
- Medical School, Tianjin University, Tianjin, 300072, China.
| | - Dong Ming
- Medical School, Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, 300072, China.
- Haihe Laboratory of Brain-Computer Interaction and Human-Machine Integration, Tianjin, 300380, China.
| |
Collapse
|
4
|
Chitolina R, Gallas-Lopes M, Reis CG, Benvenutti R, Stahlhofer-Buss T, Calcagnotto ME, Herrmann AP, Piato A. Chemically-induced epileptic seizures in zebrafish: A systematic review. Epilepsy Res 2023; 197:107236. [PMID: 37801749 DOI: 10.1016/j.eplepsyres.2023.107236] [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: 07/17/2023] [Revised: 09/14/2023] [Accepted: 09/28/2023] [Indexed: 10/08/2023]
Abstract
The use of zebrafish as a model organism is gaining evidence in the field of epilepsy as it may help to understand the mechanisms underlying epileptic seizures. As zebrafish assays became popular, the heterogeneity between protocols increased, making it hard to choose a standard protocol to conduct research while also impairing the comparison of results between studies. We conducted a systematic review to comprehensively profile the chemically-induced seizure models in zebrafish. Literature searches were performed in PubMed, Scopus, and Web of Science, followed by a two-step screening process based on inclusion/exclusion criteria. Qualitative data were extracted, and a sample of 100 studies was randomly selected for risk of bias assessment. Out of the 1058 studies identified after removing duplicates, 201 met the inclusion criteria. We found that the most common chemoconvulsants used in the reviewed studies were pentylenetetrazole (n = 180), kainic acid (n = 11), and pilocarpine (n = 10), which increase seizure severity in a dose-dependent manner. The main outcomes assessed were seizure scores and locomotion. Significant variability between the protocols was observed for administration route, duration of exposure, and dose/concentration. Of the studies subjected to risk of bias assessment, most were rated as low risk of bias for selective reporting (94%), baseline characteristics of the animals (67%), and blinded outcome assessment (54%). Randomization procedures and incomplete data were rated unclear in 81% and 68% of the studies, respectively. None of the studies reported the sample size calculation. Overall, these findings underscore the need for improved methodological and reporting practices to enhance the reproducibility and reliability of zebrafish models for studying epilepsy. Our study offers a comprehensive overview of the current state of chemically-induced seizure models in zebrafish, highlighting the common chemoconvulsants used and the variability in protocol parameters. This may be particularly valuable to researchers interested in understanding the underlying mechanisms of epileptic seizures and screening potential drug candidates in zebrafish models.
Collapse
Affiliation(s)
- Rafael Chitolina
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Psicofarmacologia e Comportamento (LAPCOM), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Matheus Gallas-Lopes
- Brazilian Reproducibility Initiative in Preclinical Systematic Review and meta-Analysis (BRISA) Collaboration, Brazil; Laboratório de Neurobiologia e Psicofarmacologia Experimental (PsychoLab), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos G Reis
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Psicofarmacologia e Comportamento (LAPCOM), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Radharani Benvenutti
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Thailana Stahlhofer-Buss
- Laboratório de Psicofarmacologia e Comportamento (LAPCOM), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Neurobiologia e Neuroquímica da Excitabilidade Neuronal e Plasticidade Sináptica (NNNESP Lab), Departamento de bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Ana P Herrmann
- Brazilian Reproducibility Initiative in Preclinical Systematic Review and meta-Analysis (BRISA) Collaboration, Brazil; Laboratório de Neurobiologia e Psicofarmacologia Experimental (PsychoLab), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Angelo Piato
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Laboratório de Psicofarmacologia e Comportamento (LAPCOM), Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
5
|
Abstract
The ability to develop effective new treatments for epilepsy may depend on improved understanding of seizure pathophysiology, about which many questions remain. Dynamic fluorescence imaging of activity at single-neuron resolution with fluorescent indicators in experimental model systems in vivo has revolutionized basic neuroscience and has the potential to do so for epilepsy research as well. Here, we review salient issues as they pertain to experimental imaging in basic epilepsy research, including commonly used imaging technologies, data processing and analysis, interpretation of results, and selected examples of how imaging-based approaches have revealed new insight into mechanisms of seizures and epilepsy.
Collapse
Affiliation(s)
- Patrick N. Lawlor
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Ethan M. Goldberg
- Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- The Epilepsy Neurogenetics Initiative, The Children’s Hospital of Philadelphia, Philadelphia
- Department of Neurology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
6
|
Aquatic Freshwater Vertebrate Models of Epilepsy Pathology: Past Discoveries and Future Directions for Therapeutic Discovery. Int J Mol Sci 2022; 23:ijms23158608. [PMID: 35955745 PMCID: PMC9368815 DOI: 10.3390/ijms23158608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 12/04/2022] Open
Abstract
Epilepsy is an international public health concern that greatly affects patients’ health and lifestyle. About 30% of patients do not respond to available therapies, making new research models important for further drug discovery. Aquatic vertebrates present a promising avenue for improved seizure drug screening and discovery. Zebrafish (Danio rerio) and African clawed frogs (Xenopus laevis and tropicalis) are increasing in popularity for seizure research due to their cost-effective housing and rearing, similar genome to humans, ease of genetic manipulation, and simplicity of drug dosing. These organisms have demonstrated utility in a variety of seizure-induction models including chemical and genetic methods. Past studies with these methods have produced promising data and generated questions for further applications of these models to promote discovery of drug-resistant seizure pathology and lead to effective treatments for these patients.
Collapse
|
7
|
Lavorato M, Nakamaru-Ogiso E, Mathew ND, Herman E, Shah NK, Haroon S, Xiao R, Seiler C, Falk MJ. Dichloroacetate improves mitochondrial function, physiology, and morphology in FBXL4 disease models. JCI Insight 2022; 7:156346. [PMID: 35881484 PMCID: PMC9462489 DOI: 10.1172/jci.insight.156346] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Pathogenic variants in the human F-box and leucine-rich repeat protein 4 (FBXL4) gene result in an autosomal recessive, multisystemic, mitochondrial disorder involving variable mitochondrial depletion and respiratory chain complex deficiencies with lactic acidemia. As no FDA-approved effective therapies for this disease exist, we sought to characterize translational C. elegans and zebrafish animal models, as well as human fibroblasts, to study FBXL4–/– disease mechanisms and identify preclinical therapeutic leads. Developmental delay, impaired fecundity and neurologic and/or muscular activity, mitochondrial dysfunction, and altered lactate metabolism were identified in fbxl-1(ok3741) C. elegans. Detailed studies of a PDHc activator, dichloroacetate (DCA), in fbxl-1(ok3741)C. elegans demonstrated its beneficial effects on fecundity, neuromotor activity, and mitochondrial function. Validation studies were performed in fbxl4sa12470 zebrafish larvae and in FBXL4–/– human fibroblasts; they showed DCA efficacy in preventing brain death, impairment of neurologic and/or muscular function, mitochondrial biochemical dysfunction, and stress-induced morphologic and ultrastructural mitochondrial defects. These data demonstrate that fbxl-1(ok3741) C. elegans and fbxl4sa12470 zebrafish provide robust translational models to study mechanisms and identify preclinical therapeutic candidates for FBXL4–/– disease. Furthermore, DCA is a lead therapeutic candidate with therapeutic benefit on diverse aspects of survival, neurologic and/or muscular function, and mitochondrial physiology that warrants rigorous clinical trial study in humans with FBXL4–/– disease.
Collapse
Affiliation(s)
- Manuela Lavorato
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Eiko Nakamaru-Ogiso
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Neal D Mathew
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Elizabeth Herman
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Nina K Shah
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Suraiya Haroon
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Rui Xiao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, United States of America
| | - Christoph Seiler
- Aquatics Core Facility, Children's Hospital of Philadelphia, Philadelphia, United States of America
| | - Marni J Falk
- Division of Human Genetics, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, United States of America
| |
Collapse
|
8
|
Mitovic N, Maksimovic S, Puflovic D, Kovacevic S, Lopicic S, Todorovic J, Spasic S, Dincic M, Ostojic JN. Cadmium significantly changes major morphometrical points and cardiovascular functional parameters during early development of zebrafish. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 87:103723. [PMID: 34391906 DOI: 10.1016/j.etap.2021.103723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 07/15/2021] [Accepted: 08/09/2021] [Indexed: 05/14/2023]
Abstract
Living organisms are commonly exposed to cadmium and other toxic metals. A vast body of research has shown the significant effects of these toxic metals on developmental processes. In order to study the role of toxic metals on early developmental stages of eukaryotes, we explored the effect of cadmium (Cd2+) contaminant on zebrafish. Thus, zebrafish embryos were exposed to 3 mg/L (16.7 μM) Cd2+ for 96 h and imaged every 24 h from the exposure onwards. Hatching rates of the eggs were determined at 72 h, followed by analyses at 96 h for: survival rate, morphometrical factors, and functional parameters of the cardiovascular system. Interestingly enough, significant hatching delays along with smaller cephalic region and some morphological abnormalities were observed in the treatment group. Moreover, substantial changes were noticed in the length of notochord and embryo, absorption of yolk sac with shorter extension, area of swimming bladder, as well as pericardium sac after Cd2+ treatment. Cadmium also caused significant abnormalities in heart physiology which could be the leading cause of mentioned morphological deformities. Herein, our results shine light on systematic acute embryological effects of cadmium in the early development of zebrafish for the first time.
Collapse
Affiliation(s)
- Nikola Mitovic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, Belgrade, Serbia.
| | - Stefan Maksimovic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Darko Puflovic
- Faculty of Electronic Engineering, University of Nis, Nis, Serbia
| | - Sanjin Kovacevic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Srdjan Lopicic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Jasna Todorovic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Svetolik Spasic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Marko Dincic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Jelena Nesovic Ostojic
- Department of Pathophysiology, Medical Faculty, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
9
|
Dworschak GC, Punetha J, Kalanithy JC, Mingardo E, Erdem HB, Akdemir ZC, Karaca E, Mitani T, Marafi D, Fatih JM, Jhangiani SN, Hunter JV, Dakal TC, Dhabhai B, Dabbagh O, Alsaif HS, Alkuraya FS, Maroofian R, Houlden H, Efthymiou S, Dominik N, Salpietro V, Sultan T, Haider S, Bibi F, Thiele H, Hoefele J, Riedhammer KM, Wagner M, Guella I, Demos M, Keren B, Buratti J, Charles P, Nava C, Héron D, Heide S, Valkanas E, Waddell LB, Jones KJ, Oates EC, Cooper ST, MacArthur D, Syrbe S, Ziegler A, Platzer K, Okur V, Chung WK, O'Shea SA, Alcalay R, Fahn S, Mark PR, Guerrini R, Vetro A, Hudson B, Schnur RE, Hoganson GE, Burton JE, McEntagart M, Lindenberg T, Yilmaz Ö, Odermatt B, Pehlivan D, Posey JE, Lupski JR, Reutter H. Biallelic and monoallelic variants in PLXNA1 are implicated in a novel neurodevelopmental disorder with variable cerebral and eye anomalies. Genet Med 2021; 23:1715-1725. [PMID: 34054129 PMCID: PMC8460429 DOI: 10.1038/s41436-021-01196-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 11/18/2022] Open
Abstract
PURPOSE To investigate the effect of PLXNA1 variants on the phenotype of patients with autosomal dominant and recessive inheritance patterns and to functionally characterize the zebrafish homologs plxna1a and plxna1b during development. METHODS We assembled ten patients from seven families with biallelic or de novo PLXNA1 variants. We describe genotype-phenotype correlations, investigated the variants by structural modeling, and used Morpholino knockdown experiments in zebrafish to characterize the embryonic role of plxna1a and plxna1b. RESULTS Shared phenotypic features among patients include global developmental delay (9/10), brain anomalies (6/10), and eye anomalies (7/10). Notably, seizures were predominantly reported in patients with monoallelic variants. Structural modeling of missense variants in PLXNA1 suggests distortion in the native protein. Our zebrafish studies enforce an embryonic role of plxna1a and plxna1b in the development of the central nervous system and the eye. CONCLUSION We propose that different biallelic and monoallelic variants in PLXNA1 result in a novel neurodevelopmental syndrome mainly comprising developmental delay, brain, and eye anomalies. We hypothesize that biallelic variants in the extracellular Plexin-A1 domains lead to impaired dimerization or lack of receptor molecules, whereas monoallelic variants in the intracellular Plexin-A1 domains might impair downstream signaling through a dominant-negative effect.
Collapse
Affiliation(s)
- Gabriel C Dworschak
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany.
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany.
- Department of Pediatrics, University Hospital Bonn, Bonn, Germany.
| | - Jaya Punetha
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jeshurun C Kalanithy
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Enrico Mingardo
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Haktan B Erdem
- Department of Medical Genetics, University of Health Sciences, Diskapi Yildirim Beyazit Training and Research Hospital, Ankara, Turkey
| | - Zeynep C Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ender Karaca
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Dana Marafi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jawid M Fatih
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shalini N Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jill V Hunter
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| | - Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Bhanupriya Dhabhai
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Omar Dabbagh
- Department of Neuroscience, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Hessa S Alsaif
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Department of Anatomy and Cell Biology, College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Reza Maroofian
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Stephanie Efthymiou
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Natalia Dominik
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Vincenzo Salpietro
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Tipu Sultan
- Department of Pediatric Neurology, Institute of Child Health, The Children's Hospital Lahore, Lahore, Pakistan
| | - Shahzad Haider
- Department of Paediatric Medicine, Wah Medical College, Rawalpindi, Pakistan
| | - Farah Bibi
- University Institute of Biochemistry & Biotechnology, PMAS - Arid Agriculture University, Rawalpindi, Pakistan
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Julia Hoefele
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Korbinian M Riedhammer
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Nephrology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matias Wagner
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Ilaria Guella
- Department of Medical Genetics, Centre for Applied Neurogenetics, University of British Columbia, Vancouver, BC, Canada
| | - Michelle Demos
- Division of Neurology, Department of Pediatrics, University of British Columbia and BC Children's Hospital, Vancouver, BC, Canada
| | - Boris Keren
- AP-HP, Hôpital Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Julien Buratti
- AP-HP, Hôpital Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Perrine Charles
- AP-HP, Hôpital Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Caroline Nava
- AP-HP, Hôpital Pitié-Salpêtrière, Département de Génétique, Paris, France
- Institut du Cerveau et de la Moelle épinière, Sorbonne Université, UMR S 1127, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Delphine Héron
- AP-HP, Hôpital Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Solveig Heide
- AP-HP, Hôpital Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Elise Valkanas
- Center for Mendelian Genomics, The Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Leigh B Waddell
- Kids Neuroscience Centre, Kids Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Kristi J Jones
- Kids Neuroscience Centre, Kids Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Emily C Oates
- Kids Neuroscience Centre, Kids Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, The University of New South Wales, Sydney, NSW, Australia
| | - Sandra T Cooper
- Kids Neuroscience Centre, Kids Research, The Children's Hospital at Westmead, Sydney, NSW, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Children's Medical Research Institute, Westmead, NSW, Australia
| | - Daniel MacArthur
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Steffen Syrbe
- Division of Pediatric Epileptology, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Andreas Ziegler
- Division of Pediatric Neurology and Metabolic Medicine, Centre for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Konrad Platzer
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Volkan Okur
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Sarah A O'Shea
- Department of Neurology, Columbia University, New York, NY, USA
| | - Roy Alcalay
- Department of Neurology, Columbia University, New York, NY, USA
| | - Stanley Fahn
- Department of Neurology, Columbia University, New York, NY, USA
| | - Paul R Mark
- Division of Medical Genetics, Helen DeVos Children's Hospital Grand Rapids, New York, MI, USA
| | - Renzo Guerrini
- Neuroscience Department, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | - Annalisa Vetro
- Neuroscience Department, Children's Hospital A. Meyer-University of Florence, Florence, Italy
| | | | | | - George E Hoganson
- Department of Pediatrics, University of Illinois, College of Medicine, Chicago, IL, USA
| | - Jennifer E Burton
- Department of Pediatrics, University of Illinois, College of Medicine, Peoria, IL, USA
| | - Meriel McEntagart
- South West Thames Regional Genetics Centre, St. George's Healthcare NHS Trust, St. George's, University of London, London, United Kingdom
| | - Tobias Lindenberg
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
| | - Öznur Yilmaz
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Benjamin Odermatt
- Institute of Anatomy and Cell Biology, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Neuroanatomy, Medical Faculty, University of Bonn, Bonn, Germany
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Section of Neurology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Texas Children's Hospital, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Heiko Reutter
- Institute of Human Genetics, Medical Faculty, University of Bonn, Bonn, Germany
- Department of Neonatology and Pediatric Intensive Care, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
10
|
Burke EA, Sturgeon M, Zastrow DB, Fernandez L, Prybol C, Marwaha S, Frothingham EP, Ward PA, Eng CM, Fresard L, Montgomery SB, Enns GM, Fisher PG, Wolfe LA, Harding B, Carrington B, Bishop K, Sood R, Huang Y, Elkahloun A, Toro C, Bassuk AG, Wheeler MT, Markello TC, Gahl WA, Malicdan MCV. Compound heterozygous KCTD7 variants in progressive myoclonus epilepsy. J Neurogenet 2021; 35:74-83. [PMID: 33970744 DOI: 10.1080/01677063.2021.1892095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
KCTD7 is a member of the potassium channel tetramerization domain-containing protein family and has been associated with progressive myoclonic epilepsy (PME), characterized by myoclonus, epilepsy, and neurological deterioration. Here we report four affected individuals from two unrelated families in which we identified KCTD7 compound heterozygous single nucleotide variants through exome sequencing. RNAseq was used to detect a non-annotated splicing junction created by a synonymous variant in the second family. Whole-cell patch-clamp analysis of neuroblastoma cells overexpressing the patients' variant alleles demonstrated aberrant potassium regulation. While all four patients experienced many of the common clinical features of PME, they also showed variable phenotypes not previously reported, including dysautonomia, brain pathology findings including a significantly reduced thalamus, and the lack of myoclonic seizures. To gain further insight into the pathogenesis of the disorder, zinc finger nucleases were used to generate kctd7 knockout zebrafish. Kctd7 homozygous mutants showed global dysregulation of gene expression and increased transcription of c-fos, which has previously been correlated with seizure activity in animal models. Together these findings expand the known phenotypic spectrum of KCTD7-associated PME, report a new animal model for future studies, and contribute valuable insights into the disease.
Collapse
Affiliation(s)
- Elizabeth A Burke
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, NIH and National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Morgan Sturgeon
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Diane B Zastrow
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | - Liliana Fernandez
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | - Cameron Prybol
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | - Shruti Marwaha
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Patricia A Ward
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Christine M Eng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Laure Fresard
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Stephen B Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Gregory M Enns
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul G Fisher
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.,Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
| | - Lynne A Wolfe
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, NIH and National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Brian Harding
- Departments of Pathology and Lab Medicine (Neuropathology), Children's Hospital of Philadelphia and the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Blake Carrington
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Kevin Bishop
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Raman Sood
- Zebrafish Core, Translational and Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Yan Huang
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, NIH and National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Abdel Elkahloun
- Microarray Core, Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Camilo Toro
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, NIH and National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | | | - Matthew T Wheeler
- Center for Undiagnosed Diseases, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas C Markello
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, NIH and National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - William A Gahl
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, NIH and National Human Genome Research Institute, NIH, Bethesda, MD, USA.,Medical Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - May Christine V Malicdan
- NIH Undiagnosed Diseases Program, Common Fund, Office of the Director, NIH and National Human Genome Research Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
11
|
Loveless R, Shay C, Teng Y. Unveiling Tumor Microenvironment Interactions Using Zebrafish Models. Front Mol Biosci 2021; 7:611847. [PMID: 33521055 PMCID: PMC7841114 DOI: 10.3389/fmolb.2020.611847] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/30/2020] [Indexed: 11/23/2022] Open
Abstract
The tumor microenvironment (TME) is a rich and active arena that is strategically evolved overtime by tumors to promote their survival and dissemination. Over the years, attention has been focused to characterize and identify the tumor-supporting roles and subsequent targeting potentials of TME components. Nevertheless, recapitulating the human TME has proved inherently challenging, leaving much to be explored. In this regard, in vivo model systems like zebrafish, with its optical clarity, ease of genetic manipulation, and high engraftment, have proven to be indispensable for TME modeling and investigation. In this review, we discuss the recent ways by which zebrafish models have lent their utility to provide new insights into the various cellular and molecular mechanisms driving TME dynamics and tumor support. Specifically, we report on innate immune cell interactions, cytokine signaling, metastatic plasticity, and other processes within the metastatic cascade. In addition, we reflect on the arrival of adult zebrafish models and the potential of patient-derived xenografts.
Collapse
Affiliation(s)
- Reid Loveless
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Chloe Shay
- Department of Pediatrics, Emory Children's Center, Emory University, Atlanta, GA, United States
| | - Yong Teng
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA, United States
- Georgia Cancer Center, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medical Laboratory, Imaging and Radiologic Sciences, College of Allied Health, Augusta University, Augusta, GA, United States
| |
Collapse
|
12
|
Gawel K, Langlois M, Martins T, van der Ent W, Tiraboschi E, Jacmin M, Crawford AD, Esguerra CV. Seizing the moment: Zebrafish epilepsy models. Neurosci Biobehav Rev 2020; 116:1-20. [PMID: 32544542 DOI: 10.1016/j.neubiorev.2020.06.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 04/20/2020] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Zebrafish are now widely accepted as a valuable animal model for a number of different central nervous system (CNS) diseases. They are suitable both for elucidating the origin of these disorders and the sequence of events culminating in their onset, and for use as a high-throughput in vivo drug screening platform. The availability of powerful and effective techniques for genome manipulation allows the rapid modelling of different genetic epilepsies and of conditions with seizures as a core symptom. With this review, we seek to summarize the current knowledge about existing epilepsy/seizures models in zebrafish (both pharmacological and genetic) and compare them with equivalent rodent and human studies. New findings obtained from the zebrafish models are highlighted. We believe that this comprehensive review will highlight the value of zebrafish as a model for investigating different aspects of epilepsy and will help researchers to use these models to their full extent.
Collapse
Affiliation(s)
- Kinga Gawel
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway; Department of Experimental and Clinical Pharmacology, Medical University of Lublin, Jaczewskiego St. 8b, 20-090, Lublin, Poland
| | | | - Teresa Martins
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg
| | - Wietske van der Ent
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway
| | - Ettore Tiraboschi
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway; Neurophysics Group, Center for Mind/Brain Sciences, University of Trento, Piazza Manifattura 1, Building 14, 38068, Rovereto, TN, Italy
| | - Maxime Jacmin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg
| | - Alexander D Crawford
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg; Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU), Oslo, Norway
| | - Camila V Esguerra
- Chemical Neuroscience Group, Centre for Molecular Medicine Norway (NCMM), University of Oslo, Gaustadalléen 21, Forskningsparken, 0349, Oslo, Norway.
| |
Collapse
|
13
|
Burrows DRW, Samarut É, Liu J, Baraban SC, Richardson MP, Meyer MP, Rosch RE. Imaging epilepsy in larval zebrafish. Eur J Paediatr Neurol 2020; 24:70-80. [PMID: 31982307 PMCID: PMC7035958 DOI: 10.1016/j.ejpn.2020.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 12/19/2022]
Abstract
Our understanding of the genetic aetiology of paediatric epilepsies has grown substantially over the last decade. However, in order to translate improved diagnostics to personalised treatments, there is an urgent need to link molecular pathophysiology in epilepsy to whole-brain dynamics in seizures. Zebrafish have emerged as a promising new animal model for epileptic seizure disorders, with particular relevance for genetic and developmental epilepsies. As a novel model organism for epilepsy research they combine key advantages: the small size of larval zebrafish allows high throughput in vivo experiments; the availability of advanced genetic tools allows targeted modification to model specific human genetic disorders (including genetic epilepsies) in a vertebrate system; and optical access to the entire central nervous system has provided the basis for advanced microscopy technologies to image structure and function in the intact larval zebrafish brain. There is a growing body of literature describing and characterising features of epileptic seizures and epilepsy in larval zebrafish. Recently genetically encoded calcium indicators have been used to investigate the neurobiological basis of these seizures with light microscopy. This approach offers a unique window into the multiscale dynamics of epileptic seizures, capturing both whole-brain dynamics and single-cell behaviour concurrently. At the same time, linking observations made using calcium imaging in the larval zebrafish brain back to an understanding of epileptic seizures largely derived from cortical electrophysiological recordings in human patients and mammalian animal models is non-trivial. In this review we briefly illustrate the state of the art of epilepsy research in zebrafish with particular focus on calcium imaging of epileptic seizures in the larval zebrafish. We illustrate the utility of a dynamic systems perspective on the epileptic brain for providing a principled approach to linking observations across species and identifying those features of brain dynamics that are most relevant to epilepsy. In the following section we survey the literature for imaging features associated with epilepsy and epileptic seizures and link these to observations made from humans and other more traditional animal models. We conclude by identifying the key challenges still facing epilepsy research in the larval zebrafish and indicate strategies for future research to address these and integrate more directly with the themes and questions that emerge from investigating epilepsy in other model systems and human patients.
Collapse
Affiliation(s)
- D R W Burrows
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - É Samarut
- Department of Neurosciences, Research Center of the University of Montreal Hospital Center, Montreal, Quebec, Canada
| | - J Liu
- Department of Neurological Surgery and Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - S C Baraban
- Department of Neurological Surgery and Weill Institute for Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - M P Richardson
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - M P Meyer
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - R E Rosch
- MRC Centre for Neurodevelopmental Disorders, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Paediatric Neurology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
14
|
Chen PY, Tu HC, Schirch V, Safo MK, Fu TF. Pyridoxamine Supplementation Effectively Reverses the Abnormal Phenotypes of Zebrafish Larvae With PNPO Deficiency. Front Pharmacol 2019; 10:1086. [PMID: 31616300 PMCID: PMC6764245 DOI: 10.3389/fphar.2019.01086] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/26/2019] [Indexed: 01/09/2023] Open
Abstract
Neonatal epileptic encephalopathy (NEE), as a result of pyridoxine 5′-phosphate oxidase (PNPO) deficiency, is a rare neural disorder characterized by intractable seizures and usually leads to early infant death. The clinical phenotypes do not respond to antiepileptic drugs but are alleviated in most cases by giving large doses of pyridoxal 5′-phosphate (PLP). PLP is the active form of vitamin B6 participating in more than 100 enzymatic pathways. One of the causes of NEE is pathogenic mutations in the gene for human PNPO (hPNPO). PNPO is a key enzyme in converting pyridoxine (PN), the common dietary form of vitamin B6, and some other B6 vitamers to PLP. More than 25 different mutations in hPNPO, which result in reduced catalytic activity, have been described for PNPO-deficiency NEE. To date, no animal model is available to test new therapeutic strategies. In this report, we describe using zebrafish with reduced activity of Pnpo as an animal model. Knocking down zPnpo resulted in developmental anomalies including brain malformation and impaired locomotor activity, similar to the clinical features of PNPO-deficiency NEE. Other anomalies include a defective circulation system. These anomalies were significantly alleviated by co-injecting either zpnpo or hPNPO mRNAs. As expected from clinical observations in humans, supplementing with PLP improved the morphological and behavioral anomalies. PN only showed marginal positive effects, and only in a few anomalies. Remarkably, pyridoxamine (PM), another dietary form of vitamin B6, showed rescue effects even at a lower concentration than PLP, presenting a possible new therapeutic treatment for PNPO-deficiency NEE. Finally, GABA, a neurotransmitter whose biosynthesis depends on a PLP-dependent enzyme, showed some positive rescue effect. These results suggest zebrafish to be a promising PNPO-deficiency model for studying PLP homeostasis and drug therapy in vivo.
Collapse
Affiliation(s)
- Po-Yuan Chen
- College of Medicine, Institute of Basic Medical Science, National Cheng Kung University, Tainan, Taiwan
| | - Hung-Chi Tu
- College of Medicine, Institute of Basic Medical Science, National Cheng Kung University, Tainan, Taiwan
| | - Verne Schirch
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| | - Martin K Safo
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, United States
| | - Tzu-Fun Fu
- College of Medicine, Institute of Basic Medical Science, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
15
|
Liu F, Du C, Tian X, Ma Y, Zhao B, Yan Y, Lin Z, Lin P, Zhou R, Wang X. A novel LGI1 missense mutation causes dysfunction in cortical neuronal migration and seizures. Brain Res 2019; 1721:146332. [PMID: 31301272 DOI: 10.1016/j.brainres.2019.146332] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/23/2019] [Accepted: 07/09/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND To explore the causative genes and pathogenesis of autosomal dominant partial epilepsy with auditory features in a large Chinese family that includes 7 patients over four generations. METHODS We used targeted exome sequencing and Sanger sequencing to validate the mutation. Zebrafish were used to explore the epileptic behavior caused by the mutation. Primary cortical neuronal culturing and in utero electroporation were used to observe the influences of the mutation on neuronal polarity and migration. RESULTS We report the identification of a novel missense mutation, c.128C > G (p. Pro43Arg), in exon 1 of LGI1. The heterozygous missense mutation, which cosegregated with the syndrome, was absent in 300 unrelated and matched-ancestor controls. The mutation inhibited the secretion of LGI1 and could not rescue the hyperactivity caused by lgi1a knockdown in zebrafish. In vitro, mutant LGI1 interrupts normal cell polarity. In agreement with these findings, dysfunctional cortical neuron migration was observed using in utero electroporation technology, which is reminiscent of the subtle structural changes in the lateral temporal region observed in the proband of this family. CONCLUSION Our findings enrich the spectrum of LGI1 mutations and support the pathogenicity of the mutation. Furthermore, additional information regarding the role of LGI1 in the development of temporal lobe epilepsy was elucidated, and a potential relationship was established between cortical neuronal migration dysfunction and autosomal dominant partial epilepsy with auditory features.
Collapse
Affiliation(s)
- Feng Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Chao Du
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Yuanlin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Bei Zhao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Yin Yan
- Department of Neurology, The Ninth People's Hospital of Chongqing, Chongqing 400016, China
| | - Zijun Lin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Peijia Lin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Ruijiao Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China
| | - Xuefeng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing 400016, China.
| |
Collapse
|
16
|
Makkar H, Verma SK, Panda PK, Jha E, Das B, Mukherjee K, Suar M. In Vivo Molecular Toxicity Profile of Dental Bioceramics in Embryonic Zebrafish ( Danio rerio). Chem Res Toxicol 2018; 31:914-923. [PMID: 30058326 DOI: 10.1021/acs.chemrestox.8b00129] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The investigation of the biocompatibility of potential and commercially available dental material is a major challenge in dental science. This study demonstrates that the zebrafish model is a novel in vivo model for investigating the biocompatibility of dental materials. Two commercially available dental materials, mineral trioxide aggregate (MTA) and Biodentine, were assessed for their biocompatibility. The biocompatibility analysis was performed in embryonic zebrafish with the help of standard toxicity assays measuring essential parameters such as survivability and hatching. The mechanistic and comparative analysis of toxicity was performed by oxidative stress analysis by measuring ROS induction and apoptosis in zebrafish exposed to dental materials at different concentrations. The molecular investigation at the protein level was done by a computational approach using in silico molecular docking and pathway analysis. The toxicity analysis showed a significant reduction in hatching and survivability rates along with morphological malformations with an increase in the concentration of exposed materials. ROS and apoptosis assay results revealed a greater biocompatibility of Biodentine as compared to that of MTA which was concentration-dependent. In silico analysis showed the significant role of the tricalcium silicate-protein ( Sod1, tp53, RUNX2B) interaction in an exhibition of toxicity. The study provides a new vision and standard in dental material sciences for assessing the biocompatibility of potential novel and commercially available dental materials.
Collapse
Affiliation(s)
| | | | - Pritam Kumar Panda
- Division of Paediatric Haematology and Oncology , University of Freiburg , Freiburg 79106 , Germany
| | - Ealisha Jha
- Department of Physics and Physical Oceanography , Memorial University of Newfoundland , St. John's , NL A1C 5S7 , Canada
| | | | - Kaushik Mukherjee
- Center for Craniofacial Molecular Biology , University of Southern California , Los Angeles , California 90089 , United States
| | | |
Collapse
|
17
|
Fuller TD, Westfall TA, Das T, Dawson DV, Slusarski DC. High-throughput behavioral assay to investigate seizure sensitivity in zebrafish implicates ZFHX3 in epilepsy. J Neurogenet 2018; 32:92-105. [PMID: 29718741 DOI: 10.1080/01677063.2018.1445247] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Epilepsy, which affects ∼1% of the population, is caused by abnormal synchronous neural activity in the central nervous system (CNS). While there is a significant genetic contribution to epilepsy, the underlying causes for the majority of genetic cases remain unknown. The NIH Undiagnosed Diseases Project (UDP) utilized exome sequencing to identify genetic variants in patients affected by various conditions with undefined etiology, including epilepsy. Confirming the functional relevance of the candidate genes identified by exome sequencing in a timely manner is crucial to translating exome data into clinically useful information. To this end, we developed a high throughput version of a seizure-sensitivity assay in zebrafish (Danio rerio) to rapidly evaluate candidate genes found by exome sequencing. We developed open access software, Studying Epilepsy In Zebrafish using R (SEIZR), to efficiently analyze the data. SEIZR was validated by disrupting function of a known epilepsy gene, prickle 1. Next, using SEIZR, we analyzed a candidate gene from the UDP screen (Zinc Finger Homeobox 3, ZFHX3), and showed that reduced ZFHX3 function in zebrafish results in a significant hyperactive response to the convulsant drug pentylenetetrazol (PTZ). We find that ZFHX3 shows strong expression in the CNS during neurogenesis including in the pallium, thalamus, tegmentum, reticular formation, and medulla oblongata - all regions which have roles in motor control and coordination. Our findings in the zebrafish confirm human ZFHX3 is a strong candidate for further neurological studies. We offer SEIZR to other researchers as a tool to rapidly and efficiently analyze large behavioral data sets.
Collapse
Affiliation(s)
- Tyson D Fuller
- a Department of Biology , University of Iowa , Iowa City , IA , USA.,b Interdisciplinary Graduate Program in Genetics , University of Iowa , Iowa City , IA , USA
| | - Trudi A Westfall
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Tirthasree Das
- a Department of Biology , University of Iowa , Iowa City , IA , USA
| | - Deborah V Dawson
- b Interdisciplinary Graduate Program in Genetics , University of Iowa , Iowa City , IA , USA.,c Iowa Institute for Oral Health Research , University of Iowa , Iowa City , IA , USA.,d Department of Biostatistics , University of Iowa , Iowa City , IA , USA
| | | |
Collapse
|
18
|
de Calbiac H, Dabacan A, Marsan E, Tostivint H, Devienne G, Ishida S, Leguern E, Baulac S, Muresan RC, Kabashi E, Ciura S. Depdc5 knockdown causes mTOR-dependent motor hyperactivity in zebrafish. Ann Clin Transl Neurol 2018; 5:510-523. [PMID: 29761115 PMCID: PMC5945968 DOI: 10.1002/acn3.542] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/18/2022] Open
Abstract
Objective DEPDC5 was identified as a major genetic cause of focal epilepsy with deleterious mutations found in a wide range of inherited forms of focal epilepsy, associated with malformation of cortical development in certain cases. Identification of frameshift, truncation, and deletion mutations implicates haploinsufficiency of DEPDC5 in the etiology of focal epilepsy. DEPDC5 is a component of the GATOR1 complex, acting as a negative regulator of mTOR signaling. Methods Zebrafish represents a vertebrate model suitable for genetic analysis and drug screening in epilepsy-related disorders. In this study, we defined the expression of depdc5 during development and established an epilepsy model with reduced Depdc5 expression. Results Here we report a zebrafish model of Depdc5 loss-of-function that displays a measurable behavioral phenotype, including hyperkinesia, circular swimming, and increased neuronal activity. These phenotypic features persisted throughout embryonic development and were significantly reduced upon treatment with the mTORC1 inhibitor, rapamycin, as well as overexpression of human WT DEPDC5 transcript. No phenotypic rescue was obtained upon expression of epilepsy-associated DEPDC5 mutations (p.Arg487* and p.Arg485Gln), indicating that these mutations cause a loss of function of the protein. Interpretation This study demonstrates that Depdc5 knockdown leads to early-onset phenotypic features related to motor and neuronal hyperactivity. Restoration of phenotypic features by WT but not epilepsy-associated Depdc5 mutants, as well as by mTORC1 inhibition confirm the role of Depdc5 in the mTORC1-dependent molecular cascades, defining this pathway as a potential therapeutic target for DEPDC5-inherited forms of focal epilepsy.
Collapse
Affiliation(s)
- Hortense de Calbiac
- Sorbonne Universités Paris VI UMR CNRS 1127 UPMC INSERM U 1127 CNRS UMR 7225 Institut du Cerveau et de la Moelle épinière - ICM Paris France.,Institut Imagine UMR Inserm 1163 University Paris Descartes Hospital Necker-Enfants Malades 24 Boulevard du Montparnasse Paris 75015 France
| | - Adriana Dabacan
- Transylvanian Institute of Neuroscience (TINS) Str. Ploiesti 33 Cluj-Napoca 400157 Romania
| | - Elise Marsan
- Sorbonne Universités Paris VI UMR CNRS 1127 UPMC INSERM U 1127 CNRS UMR 7225 Institut du Cerveau et de la Moelle épinière - ICM Paris France
| | - Hervé Tostivint
- Evolution des Régulations Endocriniennes UMR 7221 CNRS and Muséum National d'Histoire Naturelle Paris France
| | - Gabrielle Devienne
- Sorbonne Universités Paris VI UMR CNRS 1127 UPMC INSERM U 1127 CNRS UMR 7225 Institut du Cerveau et de la Moelle épinière - ICM Paris France
| | - Saeko Ishida
- Sorbonne Universités Paris VI UMR CNRS 1127 UPMC INSERM U 1127 CNRS UMR 7225 Institut du Cerveau et de la Moelle épinière - ICM Paris France
| | - Eric Leguern
- Sorbonne Universités Paris VI UMR CNRS 1127 UPMC INSERM U 1127 CNRS UMR 7225 Institut du Cerveau et de la Moelle épinière - ICM Paris France
| | - Stéphanie Baulac
- Sorbonne Universités Paris VI UMR CNRS 1127 UPMC INSERM U 1127 CNRS UMR 7225 Institut du Cerveau et de la Moelle épinière - ICM Paris France
| | - Raul C Muresan
- Transylvanian Institute of Neuroscience (TINS) Str. Ploiesti 33 Cluj-Napoca 400157 Romania
| | - Edor Kabashi
- Sorbonne Universités Paris VI UMR CNRS 1127 UPMC INSERM U 1127 CNRS UMR 7225 Institut du Cerveau et de la Moelle épinière - ICM Paris France.,Institut Imagine UMR Inserm 1163 University Paris Descartes Hospital Necker-Enfants Malades 24 Boulevard du Montparnasse Paris 75015 France
| | - Sorana Ciura
- Sorbonne Universités Paris VI UMR CNRS 1127 UPMC INSERM U 1127 CNRS UMR 7225 Institut du Cerveau et de la Moelle épinière - ICM Paris France.,Institut Imagine UMR Inserm 1163 University Paris Descartes Hospital Necker-Enfants Malades 24 Boulevard du Montparnasse Paris 75015 France
| |
Collapse
|
19
|
Xie X, Tang SC, Cai Y, Pi W, Deng L, Wu G, Chavanieu A, Teng Y. Suppression of breast cancer metastasis through the inactivation of ADP-ribosylation factor 1. Oncotarget 2018; 7:58111-58120. [PMID: 27517156 PMCID: PMC5295416 DOI: 10.18632/oncotarget.11185] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 08/05/2016] [Indexed: 12/21/2022] Open
Abstract
Metastasis is the major cause of cancer-related death in breast cancer patients, which is controlled by specific sets of genes. Targeting these genes may provide a means to delay cancer progression and allow local treatment to be more effective. We report for the first time that ADP-ribosylation factor 1 (ARF1) is the most amplified gene in ARF gene family in breast cancer, and high-level amplification of ARF1 is associated with increased mRNA expression and poor outcomes of patients with breast cancer. Knockdown of ARF1 leads to significant suppression of migration and invasion in breast cancer cells. Using the orthotopic xenograft model in NSG mice, we demonstrate that loss of ARF1 expression in breast cancer cells inhibits pulmonary metastasis. The zebrafish-metastasis model confirms that the ARF1 gene depletion suppresses breast cancer cells to metastatic disseminate throughout fish body, indicating that ARF1 is a very compelling target to limit metastasis. ARF1 function largely dependents on its activation and LM11, a cell-active inhibitor that specifically inhibits ARF1 activation through targeting the ARF1-GDP/ARNO complex at the Golgi, significantly impairs metastatic capability of breast cancer cell in zebrafish. These findings underline the importance of ARF1 in promoting metastasis and suggest that LM11 that inhibits ARF1 activation may represent a potential therapeutic approach to prevent or treat breast cancer metastasis.
Collapse
Affiliation(s)
- Xiayang Xie
- Department of Oral Biology, Augusta University, Augusta, GA, USA.,Department of Pediatrics, Emory Children's Center, Emory University, Atlanta, GA, USA
| | - Shou-Ching Tang
- Georgia Cancer Center, Augusta University, Augusta, GA, USA.,Tianjin Medical University Cancer Institute and Hospital, Tianjin, P.R. China
| | - Yafei Cai
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Wenhu Pi
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Libin Deng
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Guangyu Wu
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Alain Chavanieu
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, Université de Montpellier, CNRS, ENSCM, France
| | - Yong Teng
- Department of Oral Biology, Augusta University, Augusta, GA, USA.,Georgia Cancer Center, Augusta University, Augusta, GA, USA.,Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| |
Collapse
|
20
|
Pena IA, Roussel Y, Daniel K, Mongeon K, Johnstone D, Weinschutz Mendes H, Bosma M, Saxena V, Lepage N, Chakraborty P, Dyment DA, van Karnebeek CDM, Verhoeven-Duif N, Bui TV, Boycott KM, Ekker M, MacKenzie A. Pyridoxine-Dependent Epilepsy in Zebrafish Caused by Aldh7a1 Deficiency. Genetics 2017; 207:1501-1518. [PMID: 29061647 PMCID: PMC5714462 DOI: 10.1534/genetics.117.300137] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 10/04/2017] [Indexed: 11/25/2022] Open
Abstract
Pyridoxine-dependent epilepsy (PDE) is a rare disease characterized by mutations in the lysine degradation gene ALDH7A1 leading to recurrent neonatal seizures, which are uniquely alleviated by high doses of pyridoxine or pyridoxal 5'-phosphate (vitamin B6 vitamers). Despite treatment, neurodevelopmental disabilities are still observed in most PDE patients underlining the need for adjunct therapies. Over 60 years after the initial description of PDE, we report the first animal model for this disease: an aldh7a1-null zebrafish (Danio rerio) displaying deficient lysine metabolism and spontaneous and recurrent seizures in the larval stage (10 days postfertilization). Epileptiform electrographic activity was observed uniquely in mutants as a series of population bursts in tectal recordings. Remarkably, as is the case in human PDE, the seizures show an almost immediate sensitivity to pyridoxine and pyridoxal 5'-phosphate, with a resulting extension of the life span. Lysine supplementation aggravates the phenotype, inducing earlier seizure onset and death. By using mass spectrometry techniques, we further explored the metabolic effect of aldh7a1 knockout. Impaired lysine degradation with accumulation of PDE biomarkers, B6 deficiency, and low γ-aminobutyric acid levels were observed in the aldh7a1-/- larvae, which may play a significant role in the seizure phenotype and PDE pathogenesis. This novel model provides valuable insights into PDE pathophysiology; further research may offer new opportunities for drug discovery to control seizure activity and improve neurodevelopmental outcomes for PDE.
Collapse
Affiliation(s)
- Izabella A Pena
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ontario K1H 8L1, Canada
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Yann Roussel
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Kate Daniel
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Kevin Mongeon
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ontario K1H 8L1, Canada
| | - Devon Johnstone
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ontario K1H 8L1, Canada
| | | | - Marjolein Bosma
- Departments of Pediatrics and Clinical Genetics, Academic Medical Centre, 1105 AZ Amsterdam, The Netherlands
| | - Vishal Saxena
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Nathalie Lepage
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - Pranesh Chakraborty
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | - David A Dyment
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ontario K1H 8L1, Canada
| | - Clara D M van Karnebeek
- Departments of Pediatrics and Clinical Genetics, Academic Medical Centre, 1105 AZ Amsterdam, The Netherlands
- Department of Pediatrics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver V5Z 4H4, British Columbia, Canada
| | - Nanda Verhoeven-Duif
- Department of Genetics, Center for Molecular Medicine, University Medical Center (UMC), 3584 EA Utrecht, The Netherlands
| | - Tuan Vu Bui
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Kym M Boycott
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ontario K1H 8L1, Canada
| | - Marc Ekker
- Department of Biology, University of Ottawa, Ontario K1N 6N5, Canada
| | - Alex MacKenzie
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
- Department of Pediatrics, Faculty of Medicine, University of Ottawa, Ontario K1H 8L1, Canada
| |
Collapse
|
21
|
Bradford YM, Toro S, Ramachandran S, Ruzicka L, Howe DG, Eagle A, Kalita P, Martin R, Taylor Moxon SA, Schaper K, Westerfield M. Zebrafish Models of Human Disease: Gaining Insight into Human Disease at ZFIN. ILAR J 2017; 58:4-16. [PMID: 28838067 PMCID: PMC5886338 DOI: 10.1093/ilar/ilw040] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 12/18/2022] Open
Abstract
The Zebrafish Model Organism Database (ZFIN; https://zfin.org) is the central resource for genetic, genomic, and phenotypic data for zebrafish (Danio rerio) research. ZFIN continuously assesses trends in zebrafish research, adding new data types and providing data repositories and tools that members of the research community can use to navigate data. The many research advantages and flexibility of manipulation of zebrafish have made them an increasingly attractive animal to model and study human disease.To facilitate disease-related research, ZFIN developed support to provide human disease information as well as annotation of zebrafish models of human disease. Human disease term pages at ZFIN provide information about disease names, synonyms, and references to other databases as well as a list of publications reporting studies of human diseases in which zebrafish were used. Zebrafish orthologs of human genes that are implicated in human disease etiology are routinely studied to provide an understanding of the molecular basis of disease. Therefore, a list of human genes involved in the disease with their corresponding zebrafish ortholog is displayed on the disease page, with links to additional information regarding the genes and existing mutations. Studying human disease often requires the use of models that recapitulate some or all of the pathologies observed in human diseases. Access to information regarding existing and published models can be critical, because they provide a tractable way to gain insight into the phenotypic outcomes of the disease. ZFIN annotates zebrafish models of human disease and supports retrieval of these published models by listing zebrafish models on the disease term page as well as by providing search interfaces and data download files to access the data. The improvements ZFIN has made to annotate, display, and search data related to human disease, especially zebrafish models for disease and disease-associated gene information, should be helpful to researchers and clinicians considering the use of zebrafish to study human disease.
Collapse
Affiliation(s)
- Yvonne M. Bradford
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Sabrina Toro
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Sridhar Ramachandran
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Leyla Ruzicka
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Douglas G. Howe
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Anne Eagle
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Patrick Kalita
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Ryan Martin
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Sierra A. Taylor Moxon
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Kevin Schaper
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| | - Monte Westerfield
- Yvonne M. Bradford, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sabrina Toro, PhD, is a scientific curator for the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sridhar Ramachandran, MS, is a scientific curator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Leyla Ruzicka, PhD, is a scientific curator and senior research associate at the Zebrafish Model Organism Database, at the University of Oregon in Eugene, Oregon. Douglas G. Howe, PhD, is the Data Curation Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Anne Eagle, MSCS, is the Software Development and Project Manager at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Patrick Kalita, MS, is a software developer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Ryan Martin, MS, is a systems administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Sierra A. Taylor Moxon, BA, is the Database Administrator at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Kevin Schaper, BS, is a software engineer at the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon. Monte Westerfield, PhD, is a Professor of Biology in the Institute of Neuroscience and directs the Zebrafish Model Organism Database at the University of Oregon in Eugene, Oregon.
| |
Collapse
|
22
|
Zebrafish as a model to evaluate peptide-related cancer therapies. Amino Acids 2017; 49:1907-1913. [DOI: 10.1007/s00726-017-2388-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/27/2017] [Indexed: 02/03/2023]
|
23
|
Suvarchala G, Philip GH. Toxicity of 3,5,6-trichloro-2-pyridinol tested at multiple stages of zebrafish (Danio rerio) development. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:15515-15523. [PMID: 27121015 DOI: 10.1007/s11356-016-6684-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 04/11/2016] [Indexed: 06/05/2023]
Abstract
Organophosphate compounds (OP) are widely used throughout the world for pest control. 3,5,6-Trichloro-2-pyridinol (TCP) is a primary metabolite of two OP compounds namely CP and triclopyr. This study is carried out to know whether a metabolite of parent compound is doing well or harm to biota. The potential effect of TCP was evaluated on development as destabilization of any events transpiring during embryogenesis could be deleterious. To determine this, 4-hpf zebrafish embryos were exposed to five concentrations of TCP (200, 400, 600, 800, 1000 μg/L) or 99.5 % acetone (solvent control). Different early life-stage parameters were observed at four different developmental stages, 24, 48, 72 and 96 hpf. TCP-treated embryo/larvae showed increased mortality, delay in hatching time and decrease in percentage of hatched embryos. Reduction in heartbeat rate, blood flow and body and eye pigmentation was noticed in a dose-dependent manner. Pericardial and yolk sac edema were most severe malformations caused by TCP. Along with this crooked spine/notochord, tail deformation was noticed in hatched and unhatched embryos. The malformations observed provide a good starting point for examination of the molecular mechanisms that are affected during development by TCP. Results gain significance as TCP, which is a breakdown product, appears to be more toxic during development compared to parent compound, CP (our earlier publication).
Collapse
Affiliation(s)
- Gonuguntla Suvarchala
- Department of Zoology, Sri Krishnadevaraya University, Anantapuramu, 515003, AP, India
| | - Gundala Harold Philip
- Department of Zoology, Sri Krishnadevaraya University, Anantapuramu, 515003, AP, India.
| |
Collapse
|
24
|
Boillot M, Baulac S. Genetic models of focal epilepsies. J Neurosci Methods 2016; 260:132-43. [DOI: 10.1016/j.jneumeth.2015.06.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 06/03/2015] [Accepted: 06/04/2015] [Indexed: 01/06/2023]
|
25
|
Cunliffe VT. Building a zebrafish toolkit for investigating the pathobiology of epilepsy and identifying new treatments for epileptic seizures. J Neurosci Methods 2016. [DOI: 10.1016/j.jneumeth.2015.07.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
26
|
The promise of zebrafish as a chemical screening tool in cancer therapy. Future Med Chem 2015; 7:1395-405. [DOI: 10.4155/fmc.15.73] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cancer progression in zebrafish recapitulates many aspects of human cancer and as a result, zebrafish have been gaining popularity for their potential use in basic and translational cancer research. Human cancer can be modeled in zebrafish by induction using chemical mutagens, xenotransplantation or by genetic manipulation. Chemical screens based on zebrafish cancer models offer a rapid, powerful and inexpensive means of evaluating the potential of suppression or prevention on cancer. The identification of small molecules through such screens will serve as ideal entry points for novel chemical therapies for cancer treatment. This article outlines advances that have been made within the growing field of zebrafish cancer models and presents their advantages for chemical drug screening.
Collapse
|
27
|
Pamanji R, Bethu MS, Yashwanth B, Leelavathi S, Venkateswara Rao J. Developmental toxic effects of monocrotophos, an organophosphorous pesticide, on zebrafish (Danio rerio) embryos. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2015; 22:7744-53. [PMID: 25604565 DOI: 10.1007/s11356-015-4120-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 01/11/2015] [Indexed: 05/15/2023]
Abstract
The present study examined the response of zebrafish embryos exposed to different concentrations (10, 20, 30, 40, 50, and 60 mg/L) of monocrotophos under static conditions for 96 h. We found that mortality had occurred within 48 h at all test concentrations, later insignificant mortality was observed. Monocrotophos (MCP) can be rated as moderately toxic to the Zebrafish embryos with a 96-h median lethal concentration (LC50) of 37.44 ± 3.32 mg/L. In contrast, it greatly affected the development of zebrafish embryos by inducing several developmental abnormalities like pericardial edema, altered heart development, spinal and vertebral anomalies in a concentration-dependent manner. A significant percent reduction in length by 9-48% and heart beats by 18-51% was observed in hatchlings exposed to LC10 and LC50 concentrations at 96 h when compared to controls. The process of looping formation of heart at embryonic stage was greatly affected by the LC50 concentration of MCP. The neurotoxic potentiality of MCP was assessed by using a marker enzyme, acetylcholinesterase in both in vitro and in vivo experiments. MCP was found to be the most potent inhibitor of AChE in vitro with an IC50 value of 4.3 × 10(-4) M. The whole-body AChE enzyme activity in vivo was significantly inhibited during the exposure tenure with the maximum inhibition of 62% at 24 h.
Collapse
Affiliation(s)
- Rajesh Pamanji
- Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, 500 007, India
| | | | | | | | | |
Collapse
|
28
|
Pamanji R, Yashwanth B, Bethu MS, Leelavathi S, Ravinder K, Rao JV. Toxicity effects of profenofos on embryonic and larval development of Zebrafish (Danio rerio). ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:887-897. [PMID: 25796049 DOI: 10.1016/j.etap.2015.02.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/24/2015] [Accepted: 02/27/2015] [Indexed: 06/04/2023]
Abstract
The aim of the present study was to evaluate the developmental toxicity of profenofos to early developing Zebrafish (Danio rerio) embryos (4h post fertilization) in a static system at 1.0 to 2.25mg/L. Median lethal concentrations (LC50) of profenofos at 24-h, 48-h, 72-h and 96-h were determined as 2.04, 1.58, 1.57 and 1.56 mg/L, respectively. The hatching of embryos were recorded at every 12h interval and the median hatching time (HT50) was also calculated for each concentration. In a separate set of experiments, 96-h LC10 (0.74 mg/L) and LC50 (1.56 mg/L) concentrations were used to assess the developmental toxicity in relation to behavior, morphology, and interactions with the targeted enzyme acetylcholinesterase. Live video-microscopy revealed that the profenofos exposed embryos exhibited an abnormal development, skeletal defects and altered heart morphology in a concentration-dependent manner, which leads to alterations in the swimming behavior of hatchlings at 144-h, which indicate that developing zebrafish are sensitive to profenofos.
Collapse
Affiliation(s)
- Rajesh Pamanji
- Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - B Yashwanth
- Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - M S Bethu
- Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - S Leelavathi
- Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - K Ravinder
- Zebrafish Laboratory, Centre for Cellular and Molecular Biology, Hyderabad 500 007, India
| | - J Venkateswara Rao
- Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
| |
Collapse
|
29
|
Cunliffe VT, Baines RA, Giachello CNG, Lin WH, Morgan A, Reuber M, Russell C, Walker MC, Williams RSB. Epilepsy research methods update: Understanding the causes of epileptic seizures and identifying new treatments using non-mammalian model organisms. Seizure 2014; 24:44-51. [PMID: 25457452 DOI: 10.1016/j.seizure.2014.09.018] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 09/23/2014] [Indexed: 12/31/2022] Open
Abstract
This narrative review is intended to introduce clinicians treating epilepsy and researchers familiar with mammalian models of epilepsy to experimentally tractable, non-mammalian research models used in epilepsy research, ranging from unicellular eukaryotes to more complex multicellular organisms. The review focuses on four model organisms: the social amoeba Dictyostelium discoideum, the roundworm Caenorhabditis elegans, the fruit fly Drosophila melanogaster and the zebrafish Danio rerio. We consider recent discoveries made with each model organism and discuss the importance of these advances for the understanding and treatment of epilepsy in humans. The relative ease with which mutations in genes of interest can be produced and studied quickly and cheaply in these organisms, together with their anatomical and physiological simplicity in comparison to mammalian species, are major advantages when researchers are trying to unravel complex disease mechanisms. The short generation times of most of these model organisms also mean that they lend themselves particularly conveniently to the investigation of drug effects or epileptogenic processes across the lifecourse.
Collapse
Affiliation(s)
- Vincent T Cunliffe
- Bateson Centre, Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, United Kingdom.
| | - Richard A Baines
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom.
| | - Carlo N G Giachello
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Wei-Hsiang Lin
- Faculty of Life Sciences, University of Manchester, AV Hill Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Alan Morgan
- Department of Molecular and Cellular Physiology, Institute of Translational Medicine, University of Liverpool, Crown Street, Liverpool L69 3BX, United Kingdom.
| | - Markus Reuber
- Academic Neurology Unit, University of Sheffield, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, United Kingdom.
| | - Claire Russell
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, United Kingdom.
| | - Matthew C Walker
- Department of Clinical and Experimental Epilepsy, Institute of Neurology, University College London, Queen Square, London WC1N 3BG, United Kingdom.
| | - Robin S B Williams
- School of Biological Sciences, Royal Holloway College, University of London, Egham Hill, Egham, Surrey TW20 0EX, United Kingdom.
| |
Collapse
|
30
|
Abstract
Mutations in the LGI1 gene predispose to autosomal dominant lateral temporal lobe epilepsy, a rare hereditary form with incomplete penetrance and associated with acoustic auras. LGI1 is not a structural component of an ion channel like most epilepsy-related genes, but is a secreted protein. Mutant null mice exhibit early-onset seizures, and electrophysiological analysis shows abnormal synaptic transmission. LGI1 binds to ADAM23 on the presynaptic membrane and ADAM22 on the postsynaptic membrane, further implicating it in regulating the strength of synaptic transmission. Patients with limbic encephalitis show autoantibodies against LGI1 and develop seizures, supporting a role for LGI1 in synapse transmission in the post developmental brain. LGI1, however, also seems to be involved in aspects of neurite development and dendritic pruning, suggesting an additional role in corticogenesis. LGI1 is also involved in cell movement and suppression of dendritic outgrowth in in vitro systems, possibly involving actin cytoskeleton dynamics. Expression patterns in embryonic development correspond to areas of neuronal migration. Loss of LGI1 expression also impacts on myelination of the central and peripheral nervous systems. In zebrafish embryos, knockdown of lgi1a leads to a seizure-like behavior and abnormal brain development, providing a system to study its role in early embryogenesis. Despite being implicated in a role in both synapse transmission and neuronal development, how LGI1 predisposes to epilepsy is still largely unknown. It appears, however, that LGI1 may function differently in a cell context-specific manner, implying a complex involvement in brain development and function that remains to be defined.
Collapse
Affiliation(s)
- John K Cowell
- Georgia Regents University Cancer Center, Augusta, GA, USA.
| |
Collapse
|
31
|
Baraban SC, Löscher W. What new modeling approaches will help us identify promising drug treatments? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 813:283-94. [PMID: 25012385 DOI: 10.1007/978-94-017-8914-1_23] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite the development of numerous novel antiepileptic drugs (AEDs) in recent years, several unmet clinical needs remain, including resistance to AEDs in about 30 % of patients with epilepsy, adverse effects of AEDs that can reduce quality of life, and the lack of treatments that can prevent development of epilepsy in patients at risk. Animal models of seizures and epilepsy have been instrumental in the discovery and preclinical development of novel AEDs, but obviously the previously used models have failed to identify drugs that address unmet medical needs. Thus, we urgently need fresh ideas for improving preclinical AED development. In this review, a number of promising models will be described, including the use of simple vertebrates such as zebrafish (Danio rerio), large animal models such as the dog and newly characterized rodent models of pharmacoresistant epilepsy. While these strategies, like any animal model approach also have their limitations, they offer hope that new more effective AEDs will be identified in the coming years.
Collapse
Affiliation(s)
- Scott C Baraban
- Epilepsy Research Laboratory, Department of Neurological Surgery, University of California, San Francisco, CA, 94143, USA,
| | | |
Collapse
|
32
|
Teng Y, Xie X, Walker S, White DT, Mumm JS, Cowell JK. Evaluating human cancer cell metastasis in zebrafish. BMC Cancer 2013; 13:453. [PMID: 24089705 PMCID: PMC3852235 DOI: 10.1186/1471-2407-13-453] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/24/2013] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND In vivo metastasis assays have traditionally been performed in mice, but the process is inefficient and costly. However, since zebrafish do not develop an adaptive immune system until 14 days post-fertilization, human cancer cells can survive and metastasize when transplanted into zebrafish larvae. Despite isolated reports, there has been no systematic evaluation of the robustness of this system to date. METHODS Individual cell lines were stained with CM-Dil and injected into the perivitelline space of 2-day old zebrafish larvae. After 2-4 days fish were imaged using confocal microscopy and the number of metastatic cells was determined using Fiji software. RESULTS To determine whether zebrafish can faithfully report metastatic potential in human cancer cells, we injected a series of cells with different metastatic potential into the perivitelline space of 2 day old embryos. Using cells from breast, prostate, colon and pancreas we demonstrated that the degree of cell metastasis in fish is proportional to their invasion potential in vitro. Highly metastatic cells such as MDA231, DU145, SW620 and ASPC-1 are seen in the vasculature and throughout the body of the fish after only 24-48 hours. Importantly, cells that are not invasive in vitro such as T47D, LNCaP and HT29 do not metastasize in fish. Inactivation of JAK1/2 in fibrosarcoma cells leads to loss of invasion in vitro and metastasis in vivo, and in zebrafish these cells show limited spread throughout the zebrafish body compared with the highly metastatic parental cells. Further, knockdown of WASF3 in DU145 cells which leads to loss of invasion in vitro and metastasis in vivo also results in suppression of metastasis in zebrafish. In a cancer progression model involving normal MCF10A breast epithelial cells, the degree of invasion/metastasis in vitro and in mice is mirrored in zebrafish. Using a modified version of Fiji software, it is possible to quantify individual metastatic cells in the transparent larvae to correlate with invasion potential. We also demonstrate, using lung cancers, that the zebrafish model can evaluate the metastatic ability of cancer cells isolated from primary tumors. CONCLUSIONS The zebrafish model described here offers a rapid, robust, and inexpensive means of evaluating the metastatic potential of human cancer cells. Using this model it is possible to critically evaluate whether genetic manipulation of signaling pathways affects metastasis and whether primary tumors contain metastatic cells.
Collapse
Affiliation(s)
- Yong Teng
- Cancer Center, Georgia Regents University, Augusta, GA, USA
| | - Xiayang Xie
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, USA
- Vision Discovery Institute, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA
| | - Steven Walker
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, USA
- Vision Discovery Institute, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA
| | - David T White
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, USA
- Vision Discovery Institute, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA
| | - Jeff S Mumm
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, USA
- Vision Discovery Institute, Georgia Regents University, 1120 15th Street, Augusta, GA 30912, USA
| | - John K Cowell
- Cancer Center, Georgia Regents University, Augusta, GA, USA
| |
Collapse
|
33
|
Lyons PJ, Sapio MR, Fricker LD. Zebrafish cytosolic carboxypeptidases 1 and 5 are essential for embryonic development. J Biol Chem 2013; 288:30454-30462. [PMID: 24022483 DOI: 10.1074/jbc.m113.497933] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The cytosolic carboxypeptidases (CCPs) are a subfamily of metalloenzymes within the larger M14 family of carboxypeptidases that have been implicated in the post-translational modification of tubulin. It has been suggested that at least four of the six mammalian CCPs function as tubulin deglutamylases. However, it is not yet clear whether these enzymes play redundant or unique roles within the cell. To address this question, genes encoding CCPs were identified in the zebrafish genome. Analysis by quantitative polymerase chain reaction indicated that CCP1, CCP2, CCP5, and CCP6 mRNAs were detectable between 2 h and 8 days postfertilization with highest levels 5-8 days postfertilization. CCP1, CCP2, and CCP5 mRNAs were predominantly expressed in tissues such as the brain, olfactory placodes, and pronephric ducts. Morpholino oligonucleotide-mediated knockdown of CCP1 and CCP5 mRNA resulted in a common phenotype including ventral body curvature and hydrocephalus. Confocal microscopy of morphant zebrafish revealed olfactory placodes with defective morphology as well as pronephric ducts with increased polyglutamylation. These data suggest that CCP1 and CCP5 play important roles in developmental processes, particularly the development and functioning of cilia. The robust and similar defects upon knockdown suggest that each CCP may have a function in microtubule modification and ciliary function and that other CCPs are not able to compensate for the loss of one.
Collapse
Affiliation(s)
| | - Matthew R Sapio
- Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | | |
Collapse
|
34
|
Niederriter AR, Davis EE, Golzio C, Oh EC, Tsai IC, Katsanis N. In vivo modeling of the morbid human genome using Danio rerio. J Vis Exp 2013:e50338. [PMID: 23995499 DOI: 10.3791/50338] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Here, we present methods for the development of assays to query potentially clinically significant nonsynonymous changes using in vivo complementation in zebrafish. Zebrafish (Danio rerio) are a useful animal system due to their experimental tractability; embryos are transparent to enable facile viewing, undergo rapid development ex vivo, and can be genetically manipulated. These aspects have allowed for significant advances in the analysis of embryogenesis, molecular processes, and morphogenetic signaling. Taken together, the advantages of this vertebrate model make zebrafish highly amenable to modeling the developmental defects in pediatric disease, and in some cases, adult-onset disorders. Because the zebrafish genome is highly conserved with that of humans (~70% orthologous), it is possible to recapitulate human disease states in zebrafish. This is accomplished either through the injection of mutant human mRNA to induce dominant negative or gain of function alleles, or utilization of morpholino (MO) antisense oligonucleotides to suppress genes to mimic loss of function variants. Through complementation of MO-induced phenotypes with capped human mRNA, our approach enables the interpretation of the deleterious effect of mutations on human protein sequence based on the ability of mutant mRNA to rescue a measurable, physiologically relevant phenotype. Modeling of the human disease alleles occurs through microinjection of zebrafish embryos with MO and/or human mRNA at the 1-4 cell stage, and phenotyping up to seven days post fertilization (dpf). This general strategy can be extended to a wide range of disease phenotypes, as demonstrated in the following protocol. We present our established models for morphogenetic signaling, craniofacial, cardiac, vascular integrity, renal function, and skeletal muscle disorder phenotypes, as well as others.
Collapse
Affiliation(s)
- Adrienne R Niederriter
- Center for Human Disease Modeling, Department of Cell Biology, Duke University Medical Center, USA
| | | | | | | | | | | |
Collapse
|
35
|
Abstract
The development and function of the vertebrate nervous system depend on specific interactions between different cell types. Two examples of such interactions are synaptic transmission and myelination. LGI1-4 (leucine-rich glioma inactivated proteins) play important roles in these processes. They are secreted proteins consisting of an LRR (leucine-rich repeat) domain and a so-called epilepsy-associated or EPTP (epitempin) domain. Both domains are thought to function in protein–protein interactions. The first LGI gene to be identified, LGI1, was found at a chromosomal translocation breakpoint in a glioma cell line. It was subsequently found mutated in ADLTE (autosomal dominant lateral temporal (lobe) epilepsy) also referred to as ADPEAF (autosomal dominant partial epilepsy with auditory features). LGI1 protein appears to act at synapses and antibodies against LGI1 may cause the autoimmune disorder limbic encephalitis. A similar function in synaptic remodelling has been suggested for LGI2, which is mutated in canine Benign Familial Juvenile Epilepsy. LGI4 is required for proliferation of glia in the peripheral nervous system and binds to a neuronal receptor, ADAM22, to foster ensheathment and myelination of axons by Schwann cells. Thus, LGI proteins play crucial roles in nervous system development and function and their study is highly important, both to understand their biological functions and for their therapeutic potential. Here, we review our current knowledge about this important family of proteins, and the progress made towards understanding their functions.
Collapse
|
36
|
Mussulini BHM, Leite CE, Zenki KC, Moro L, Baggio S, Rico EP, Rosemberg DB, Dias RD, Souza TM, Calcagnotto ME, Campos MM, Battastini AM, de Oliveira DL. Seizures induced by pentylenetetrazole in the adult zebrafish: a detailed behavioral characterization. PLoS One 2013; 8:e54515. [PMID: 23349914 PMCID: PMC3549980 DOI: 10.1371/journal.pone.0054515] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 12/12/2012] [Indexed: 11/23/2022] Open
Abstract
Pentylenetetrazole (PTZ) is a common convulsant agent used in animal models to investigate the mechanisms of seizures. Although adult zebrafish have been recently used to study epileptic seizures, a thorough characterization of the PTZ-induced seizures in this animal model is missing. The goal of this study was to perform a detailed temporal behavior profile characterization of PTZ-induced seizure in adult zebrafish. The behavioral profile during 20 min of PTZ immersion (5, 7.5, 10, and 15 mM) was characterized by stages defined as scores: (0) short swim, (1) increased swimming activity and high frequency of opercular movement, (2) erratic movements, (3) circular movements, (4) clonic seizure-like behavior, (5) fall to the bottom of the tank and tonic seizure-like behavior, (6) death. Animals exposed to distinct PTZ concentrations presented different seizure profiles, intensities and latencies to reach all scores. Only animals immersed into 15 mM PTZ showed an increased time to return to the normal behavior (score 0), after exposure. Total mortality rate at 10 and 15 mM were 33% and 50%, respectively. Considering all behavioral parameters, 5, 7.5, 10, and 15 mM PTZ, induced seizures with low, intermediate, and high severity, respectively. Pretreatment with diazepam (DZP) significantly attenuated seizure severity. Finally, the brain PTZ levels in adult zebrafish immersed into the chemoconvulsant solution at 5 and 10 mM were comparable to those described for the rodent model, with a peak after a 20-min of exposure. The PTZ brain levels observed after 2.5-min PTZ exposure and after 60-min removal from exposure were similar. Altogether, our results showed a detailed temporal behavioral characterization of a PTZ epileptic seizure model in adult zebrafish. These behavioral analyses and the simple method for PTZ quantification could be considered as important tools for future investigations and translational research.
Collapse
Affiliation(s)
- Ben Hur M. Mussulini
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- * E-mail: (BHMM) (BM); (DLdO) (Dd)
| | - Carlos E. Leite
- Instituto de Toxicologia e Farmacologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Kamila C. Zenki
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Luana Moro
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Suelen Baggio
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo P. Rico
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Denis B. Rosemberg
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Renato D. Dias
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Tadeu M. Souza
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Maria E. Calcagnotto
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Maria M. Campos
- Instituto de Toxicologia e Farmacologia, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ana M. Battastini
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Diogo L. de Oliveira
- Departamento de Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- * E-mail: (BHMM) (BM); (DLdO) (Dd)
| |
Collapse
|
37
|
Mei X, Wu S, Bassuk AG, Slusarski DC. Mechanisms of prickle1a function in zebrafish epilepsy and retinal neurogenesis. Dis Model Mech 2013; 6:679-88. [PMID: 23324328 PMCID: PMC3634651 DOI: 10.1242/dmm.010793] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Epilepsy is a complex neurological disorder characterized by unprovoked seizures. The etiology is heterogeneous with both genetic and environmental causes. Genes that regulate neurotransmitters and ion channels in the central nervous system have been associated with epilepsy. However, a recent screening in human epilepsy patients identified mutations in the PRICKLE1 (PK1) locus, highlighting a potentially novel mechanism underlying seizures. PK1 is a core component of the planar cell polarity network that regulates tissue polarity. Zebrafish studies have shown that Pk1 coordinates cell movement, neuronal migration and axonal outgrowth during embryonic development. Yet how dysfunction of Pk1 relates to epilepsy is unknown. To address the mechanism underlying epileptogenesis, we used zebrafish to characterize Pk1a function and epilepsy-related mutant forms. We show that knockdown of pk1a activity sensitizes zebrafish larva to a convulsant drug. To model defects in the central nervous system, we used the retina and found that pk1a knockdown induces neurite outgrowth defects; yet visual function is maintained. Furthermore, we characterized the functional and biochemical properties of the PK1 mutant forms identified in human patients. Functional analyses demonstrate that the wild-type Pk1a partially suppresses the gene knockdown retinal defects but not the mutant forms. Biochemical analysis reveals increased ubiquitylation of one mutant form and decreased translational efficiency of another mutant form compared with the wild-type Pk1a. Taken together, our results indicate that mutation of human PK1 could lead to defects in neurodevelopment and signal processing, providing insight into seizure predisposition in these patients.
Collapse
Affiliation(s)
- Xue Mei
- Department of Biology, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | |
Collapse
|
38
|
Xie X, Mathias JR, Smith MA, Walker SL, Teng Y, Distel M, Köster RW, Sirotkin HI, Saxena MT, Mumm JS. Silencer-delimited transgenesis: NRSE/RE1 sequences promote neural-specific transgene expression in a NRSF/REST-dependent manner. BMC Biol 2012. [PMID: 23198762 PMCID: PMC3529185 DOI: 10.1186/1741-7007-10-93] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We have investigated a simple strategy for enhancing transgene expression specificity by leveraging genetic silencer elements. The approach serves to restrict transgene expression to a tissue of interest - the nervous system in the example provided here - thereby promoting specific/exclusive targeting of discrete cellular subtypes. Recent innovations are bringing us closer to understanding how the brain is organized, how neural circuits function, and how neurons can be regenerated. Fluorescent proteins enable mapping of the 'connectome', optogenetic tools allow excitable cells to be short-circuited or hyperactivated, and targeted ablation of neuronal subtypes facilitates investigations of circuit function and neuronal regeneration. Optimally, such toolsets need to be expressed solely within the cell types of interest as off-site expression makes establishing causal relationships difficult. To address this, we have exploited a gene 'silencing' system that promotes neuronal specificity by repressing expression in non-neural tissues. This methodology solves non-specific background issues that plague large-scale enhancer trap efforts and may provide a means of leveraging promoters/enhancers that otherwise express too broadly to be of value for in vivo manipulations. RESULTS We show that a conserved neuron-restrictive silencer element (NRSE) can function to restrict transgene expression to the nervous system. The neuron-restrictive silencing factor/repressor element 1 silencing transcription factor (NRSF/REST) transcriptional repressor binds NRSE/repressor element 1 (RE1) sites and silences gene expression in non-neuronal cells. Inserting NRSE sites into transgenes strongly biased expression to neural tissues. NRSE sequences were effective in restricting expression of bipartite Gal4-based 'driver' transgenes within the context of an enhancer trap and when associated with a defined promoter and enhancer. However, NRSE sequences did not serve to restrict expression of an upstream activating sequence (UAS)-based reporter/effector transgene when associated solely with the UAS element. Morpholino knockdown assays showed that NRSF/REST expression is required for NRSE-based transgene silencing. CONCLUSIONS Our findings demonstrate that the addition of NRSE sequences to transgenes can provide useful new tools for functional studies of the nervous system. However, the general approach may be more broadly applicable; tissue-specific silencer elements are operable in tissues other than the nervous system, suggesting this approach can be similarly applied to other paradigms. Thus, creating synthetic associations between endogenous regulatory elements and tissue-specific silencers may facilitate targeting of cellular subtypes for which defined promoters/enhancers are lacking.
Collapse
Affiliation(s)
- Xiayang Xie
- Department of Cellular Biology and Anatomy, Georgia Health Sciences University, Augusta, GA 30912, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Braida D, Donzelli A, Martucci R, Ponzoni L, Pauletti A, Sala M. Neurohypophyseal hormones protect against pentylenetetrazole-induced seizures in zebrafish: role of oxytocin-like and V1a-like receptor. Peptides 2012; 37:327-33. [PMID: 22828174 DOI: 10.1016/j.peptides.2012.07.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 07/16/2012] [Accepted: 07/16/2012] [Indexed: 02/04/2023]
Abstract
Oxytocin (OT) and arginine-vasopressin (AVP) are involved in the physiological response to different stressors like the occurrence of seizures which is regarded as a severe stress factor. Zebrafish (Danio rerio) is recently featured as a model of epilepsy but the role of neurohypophyseal hormones on this teleost is still unknown. We attempted to determine whether non-mammalian homologues like isotocin (IT) and vasotocin (AVT) affected pentylenetetrazole (PTZ)-induced seizures in adult zebrafish in comparison with OT/AVP. The mechanism was studied using the most selective OT and AVP receptor antagonists. Zebrafish were injected i.m. with increasing doses (0.1-40 ng/kg) of the neuropeptides 10 min before PTZ exposure. DesGly-NH2-d(CH2)5-[D-Tyr2,Thr4]OVT (desglyDTyrOVT) for OT receptor and SR49059 for V1a subtype receptor, were injected together with each agonist 20 min before PTZ exposure. All the peptides significantly decreased the number of seizures, increased the mean latency time to the first seizure and decreased lethality. This protective effect led to a dose-response curve following a U-shaped form. IT was approximately 40 times more active than OT while AVT was 20 times more potent than AVP in reducing the number of seizures. DesglyDTyrOVT was more effective in antagonizing OT/IT, while SR49059 mainly blocked AVP/AVT-induced protection against PTZ-induced seizures. The present findings provide direct evidence of an important involvement of IT/OT and AVP/AVT as anticonvulsant agents against PTZ-induced seizures with a receptor-mediated mechanism in zebrafish. These data reinforce zebrafish as an emerging experimental model to study and identify new antiepileptic drugs.
Collapse
Affiliation(s)
- Daniela Braida
- Dipartimento di Biotecnologie mediche e Medicina traslazionale, Università degli Studi di Milano, Via Vanvitelli 32, Milan, Italy
| | | | | | | | | | | |
Collapse
|