151
|
Konno K, Kobayashi-Sun J, Arai F, Kobayashi I, Sugiyama D. Hematopoietic Cell Isolation by Antibody-Free Flow Cytometry in the Zebrafish Embryo. Methods Mol Biol 2022; 2520:171-180. [PMID: 35575971 DOI: 10.1007/7651_2021_459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The zebrafish is a useful model to identify genes functioning in hematopoiesis, owing to high conservation of hematopoiesis. Flow cytometry is widely used to isolate and quantitatively characterize human and mouse hematopoietic cells, often using fluorescently labeled antibodies. However, such analysis is limited in zebrafish due to lack of antibodies that recognize zebrafish hematopoietic cells. We here describe methods for isolation of hematopoietic cells by antibody-free flow cytometry in the zebrafish embryo. Hematopoietic stem cells (HSCs) are specified from a specific subset of vascular endothelial cells, the hemogenic endothelial cell (HEC), by a high level of Notch signaling. In combination with a Notch reporter line (Tp1:GFP) and a vascular specific line (fli1a:dsRed), HECs can be isolated as Tp1:GFPhigh fli1a:dsRed+ cells at 20-22 hours post-fertilization (hpf). Zebrafish erythrocytes can be purified using 1,5-bis{[2-(dimethylamino)ethyl]amino}-4, 8-dihydroxyanthracene-9,10-dione (DRAQ5), a DNA-staining fluorescent dye, and gata1:dsRed (erythroid marker line). DRAQ5high dsRed+ cells are morphologically erythrocyte-like, hemoglobin-stained positive, and express erythropoiesis-related genes. Zebrafish neutrophils can be also isolated using the lectin Phaseolus vulgaris erythroagglutinin (PHA-E) and DRAQ5. PHA-Elow DRAQ5low cells have myeloperoxidase activity, are Sudan Black B-positive, and express neutrophil-related genes. These methods will help to perform genetical and functional assays for various types of hematopoietic cells in zebrafish embryos.
Collapse
Affiliation(s)
- Katsuhiro Konno
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Translational Research Center, Hiroshima University, Hiroshima, Japan
| | - Jingjing Kobayashi-Sun
- Division of Life Sciences, Graduate School of Natural Science and Technology, Kanazawa University, Ishikawa, Japan
| | - Fumio Arai
- Department of Stem Cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Isao Kobayashi
- Faculty of Biological Science and Technology, Institute of Science and Engineering, Kanazawa University, Ishikawa, Japan
| | - Daisuke Sugiyama
- Translational Research Center, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
152
|
Weiss JM, Lumaquin-Yin D, Montal E, Suresh S, Leonhardt CS, White RM. Shifting the focus of zebrafish toward a model of the tumor microenvironment. eLife 2022; 11:69703. [PMID: 36538362 PMCID: PMC9767465 DOI: 10.7554/elife.69703] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/21/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer cells exist in a complex ecosystem with numerous other cell types in the tumor microenvironment (TME). The composition of this tumor/TME ecosystem will vary at each anatomic site and affects phenotypes such as initiation, metastasis, and drug resistance. A mechanistic understanding of the large number of cell-cell interactions between tumor and TME requires models that allow us to both characterize as well as genetically perturb this complexity. Zebrafish are a model system optimized for this problem, because of the large number of existing cell-type-specific drivers that can label nearly any cell in the TME. These include stromal cells, immune cells, and tissue resident normal cells. These cell-type-specific promoters/enhancers can be used to drive fluorophores to facilitate imaging and also CRISPR cassettes to facilitate perturbations. A major advantage of the zebrafish is the ease by which large numbers of TME cell types can be studied at once, within the same animal. While these features make the zebrafish well suited to investigate the TME, the model has important limitations, which we also discuss. In this review, we describe the existing toolset for studying the TME using zebrafish models of cancer and highlight unique biological insights that can be gained by leveraging this powerful resource.
Collapse
Affiliation(s)
- Joshua M Weiss
- Weill-Cornel Medical College, Tri-Institutional M.D./Ph.D. ProgramNew YorkUnited States
| | - Dianne Lumaquin-Yin
- Weill-Cornel Medical College, Tri-Institutional M.D./Ph.D. ProgramNew YorkUnited States
| | - Emily Montal
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Shruthy Suresh
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Carl S Leonhardt
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States
| | - Richard M White
- Memorial Sloan Kettering Cancer Center, Department of Cancer Biology & GeneticsNew YorkUnited States,Department of Medicine, Memorial Sloan Kettering Cancer CenterNew YorkUnited States
| |
Collapse
|
153
|
Bu H, Ding Y, Li J, Zhu P, Shih YH, Wang M, Zhang Y, Lin X, Xu X. Inhibition of mTOR or MAPK ameliorates vmhcl/myh7 cardiomyopathy in zebrafish. JCI Insight 2021; 6:154215. [PMID: 34935644 PMCID: PMC8783688 DOI: 10.1172/jci.insight.154215] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/03/2021] [Indexed: 01/25/2023] Open
Abstract
Myosin heavy chain 7 (MYH7) is a major causative gene for hypertrophic cardiomyopathy, but the affected signaling pathways and therapeutics remain elusive. In this research, we identified ventricle myosin heavy chain like (vmhcl) as a zebrafish homolog of human MYH7, and we generated vmhcl frameshift mutants. We noted vmhcl-based embryonic cardiac dysfunction (VEC) in the vmhcl homozygous mutants and vmhcl-based adult cardiomyopathy (VAC) phenotypes in the vmhcl heterozygous mutants. Using the VEC model, we assessed 7 known cardiomyopathy signaling pathways pharmacologically and 11 candidate genes genetically via CRISPR/Cas9 genome editing technology based on microhomology-mediated end joining (MMEJ). Both studies converged on therapeutic benefits of mTOR or mitogen-activated protein kinase (MAPK) inhibition of VEC. While mTOR inhibition rescued the enlarged nuclear size of cardiomyocytes, MAPK inhibition restored the prolonged cell shape in the VEC model. The therapeutic effects of mTOR and MAPK inhibition were later validated in the VAC model. Together, vmhcl/myh7 loss of function is sufficient to induce cardiomyopathy in zebrafish. The VEC and VAC models in zebrafish are amenable to both efficient genetic and chemical genetic tools, offering a rapid in vivo platform for discovering candidate signaling pathways of MYH7 cardiomyopathy.
Collapse
Affiliation(s)
- Haisong Bu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiothoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yonghe Ding
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jiarong Li
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Yu-Huan Shih
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Mingmin Wang
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuji Zhang
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Xueying Lin
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
154
|
Single-cell transcriptomic analysis of zebrafish cranial neural crest reveals spatiotemporal regulation of lineage decisions during development. Cell Rep 2021; 37:110140. [PMID: 34936864 PMCID: PMC8741273 DOI: 10.1016/j.celrep.2021.110140] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/28/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022] Open
Abstract
Neural crest (NC) cells migrate throughout vertebrate embryos to give rise to a huge variety of cell types, but when and where lineages emerge and their regulation remain unclear. We have performed single-cell RNA sequencing (RNA-seq) of cranial NC cells from the first pharyngeal arch in zebrafish over several stages during migration. Computational analysis combining pseudotime and real-time data reveals that these NC cells first adopt a transitional state, becoming specified mid-migration, with the first lineage decisions being skeletal and pigment, followed by neural and glial progenitors. In addition, by computationally integrating these data with RNA-seq data from a transgenic Wnt reporter line, we identify gene cohorts with similar temporal responses to Wnts during migration and show that one, Atp6ap2, is required for melanocyte differentiation. Together, our results show that cranial NC cell lineages arise progressively and uncover a series of spatially restricted cell interactions likely to regulate such cell-fate decisions. Tatarakis et al. provide a single-cell transcriptomic timeline of cranial neural crest (NC) development in zebrafish and address long-standing questions surrounding the integration of NC cell migration and lineage specification. They find that lineages are specified mid-migration. These fate decisions correspond to shifts in Wnt signaling, and lineages rapidly segregate.
Collapse
|
155
|
In vivo identification and validation of novel potential predictors for human cardiovascular diseases. PLoS One 2021; 16:e0261572. [PMID: 34919578 PMCID: PMC8682894 DOI: 10.1371/journal.pone.0261572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 12/03/2021] [Indexed: 12/30/2022] Open
Abstract
Genetics crucially contributes to cardiovascular diseases (CVDs), the global leading cause of death. Since the majority of CVDs can be prevented by early intervention there is a high demand for the identification of predictive causative genes. While genome wide association studies (GWAS) correlate genes and CVDs after diagnosis and provide a valuable resource for such causative candidate genes, often preferentially those with previously known or suspected function are addressed further. To tackle the unaddressed blind spot of understudied genes, we particularly focused on the validation of human heart phenotype-associated GWAS candidates with little or no apparent connection to cardiac function. Building on the conservation of basic heart function and underlying genetics from fish to human we combined CRISPR/Cas9 genome editing of the orthologs of human GWAS candidates in isogenic medaka with automated high-throughput heart rate analysis. Our functional analyses of understudied human candidates uncovered a prominent fraction of heart rate associated genes from adult human patients impacting on the heart rate in embryonic medaka already in the injected generation. Following this pipeline, we identified 16 GWAS candidates with potential diagnostic and predictive power for human CVDs.
Collapse
|
156
|
Tromp A, Robinson K, Hall TE, Mowry B, Giacomotto J. Pipeline for generating stable large genomic deletions in zebrafish, from small domains to whole gene excisions. G3 (BETHESDA, MD.) 2021; 11:jkab321. [PMID: 34499171 PMCID: PMC8664487 DOI: 10.1093/g3journal/jkab321] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 08/27/2021] [Indexed: 11/14/2022]
Abstract
Here we describe a short feasibility study and methodological framework for the production of stable, CRISPR/Cas9-based, large genomic deletions in zebrafish, ranging from several base pairs (bp) to hundreds of kilobases (kb). Using a cocktail of four single guide RNAs (sgRNAs) targeting a single genomic region mixed with a marker-sgRNA against the pigmentation gene tyrosinase, we demonstrate that one can easily and accurately excise genomic regions such as promoters, protein domains, specific exons, or whole genes. We exemplify this technique with a complex gene family, neurexins, composed of three duplicated genes with multiple promoters and intricate splicing processes leading to thousands of isoforms. We precisely deleted small regions such as their transmembrane domains (150 bp deletion in average) to their entire genomic locus (300 kb deletion for nrxn1a for instance). We find that both the concentration and ratio of Cas9/sgRNAs are critical for the successful generation of these large deletions and, interestingly, that in our study, their transmission frequency does not seem to decrease with increasing distance between sgRNA target sites. Considering the growing reports and debate about genetically compensated small indel mutants, the use of large-deletion approaches is likely to be widely adopted in studies of gene function. This strategy will also be key to the study of non-coding genomic regions. Note that we are also describing here a custom method to produce the sgRNAs, which proved to be faster and more robust than the ones traditionally used in the community to date.
Collapse
Affiliation(s)
- Alisha Tromp
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Kate Robinson
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Thomas E Hall
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Bryan Mowry
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
- Queensland Centre for Mental Health Research, West Moreton Hospital and Health Service, Wacol, QLD 4076 Australia
| | - Jean Giacomotto
- Queensland Brain Institute, The University of Queensland, St Lucia, Queensland 4072, Australia
- Queensland Centre for Mental Health Research, West Moreton Hospital and Health Service, Wacol, QLD 4076 Australia
| |
Collapse
|
157
|
Homeostatic Regulation of Glucocorticoid Receptor Activity by Hypoxia-Inducible Factor 1: From Physiology to Clinic. Cells 2021; 10:cells10123441. [PMID: 34943949 PMCID: PMC8699886 DOI: 10.3390/cells10123441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/02/2021] [Accepted: 12/04/2021] [Indexed: 11/16/2022] Open
Abstract
Glucocorticoids (GCs) represent a well-known class of lipophilic steroid hormones biosynthesised, with a circadian rhythm, by the adrenal glands in humans and by the inter-renal tissue in teleost fish (e.g., zebrafish). GCs play a key role in the regulation of numerous physiological processes, including inflammation, glucose, lipid, protein metabolism and stress response. This is achieved through binding to their cognate receptor, GR, which functions as a ligand-activated transcription factor. Due to their potent anti-inflammatory and immune-suppressive action, synthetic GCs are broadly used for treating pathological disorders that are very often linked to hypoxia (e.g., rheumatoid arthritis, inflammatory, allergic, infectious, and autoimmune diseases, among others) as well as to prevent graft rejections and against immune system malignancies. However, due to the presence of adverse effects and GC resistance their therapeutic benefits are limited in patients chronically treated with steroids. For this reason, understanding how to fine-tune GR activity is crucial in the search for novel therapeutic strategies aimed at reducing GC-related side effects and effectively restoring homeostasis. Recent research has uncovered novel mechanisms that inhibit GR function, thereby causing glucocorticoid resistance, and has produced some surprising new findings. In this review we analyse these mechanisms and focus on the crosstalk between GR and HIF signalling. Indeed, its comprehension may provide new routes to develop novel therapeutic targets for effectively treating immune and inflammatory response and to simultaneously facilitate the development of innovative GCs with a better benefits-risk ratio.
Collapse
|
158
|
Xia Z, Bi X, Yang S, Yang X, Song Z, Wei J, Xu P, Rink L, Min J, Wang F. Metal transporter Slc30a1 controls pharyngeal neural crest differentiation via the zinc-Snai2-Jag1 cascade. MedComm (Beijing) 2021; 2:778-797. [PMID: 34977877 PMCID: PMC8706747 DOI: 10.1002/mco2.91] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
The pharyngeal arch (PA) is a neural crest (NC)-derived organ that is transiently developed during embryogenesis and is required for the subsequent development of various tissues. However, the role of zinc during PA differentiation from NC progenitor cells is unknown. Here, we found that the metal transporters Slc30a1a and Slc30a1b mediate zinc homeostasis during PA differentiation. Slc30a1-deficient zebrafish develop zinc accumulation in NC cells, with increased expression of stemness markers and PA dorsal genes, and SMART-seq analyses revealed that the genes snai2 and jag1b may serve as downstream targets. Furthermore, functional studies showed that knocking down either snai2 or jag1b rescues PA development in Slc30a1-deficient zebrafish. Notably, we identified the double zinc-finger domain in the transcription factor Snai2 as a zinc-responsive element that regulates jag1b expression. Our findings indicate that the Slc30a1/zinc-snai2-jag1b axis is an essential regulatory network controlling PA differentiation, shedding new light on the function of zinc homeostasis in maintaining NC cell stemness and multipotency in vertebrates.
Collapse
Affiliation(s)
- Zhidan Xia
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Xinying Bi
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
- The First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangChina
| | - Sisi Yang
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Xiu Yang
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Zijun Song
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Jiayu Wei
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Pengfei Xu
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Lothar Rink
- Faculty of MedicineInstitute of ImmunologyRWTH Aachen UniversityAachenGermany
| | - Junxia Min
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
| | - Fudi Wang
- The First Affiliated HospitalSchool of Public HealthInstitute of Translational MedicineInstitute of GeneticsZhejiang University School of MedicineHangzhouChina
- The First Affiliated HospitalHengyang Medical SchoolUniversity of South ChinaHengyangChina
| |
Collapse
|
159
|
Samper-Martín B, Sarrias A, Lázaro B, Pérez-Montero M, Rodríguez-Rodríguez R, Ribeiro MPC, Bañón A, Wolfgeher D, Jessen HJ, Alsina B, Clotet J, Kron SJ, Saiardi A, Jiménez J, Bru S. Polyphosphate degradation by Nudt3-Zn 2+ mediates oxidative stress response. Cell Rep 2021; 37:110004. [PMID: 34788624 DOI: 10.1016/j.celrep.2021.110004] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 10/08/2021] [Accepted: 10/22/2021] [Indexed: 02/06/2023] Open
Abstract
Polyphosphate (polyP) is a polymer of hundreds of phosphate residues present in all organisms. In mammals, polyP is involved in crucial physiological processes, including coagulation, inflammation, and stress response. However, after decades of research, the metabolic enzymes are still unknown. Here, we purify and identify Nudt3, a NUDIX family member, as the enzyme responsible for polyP phosphatase activity in mammalian cells. We show that Nudt3 shifts its substrate specificity depending on the cation; specifically, Nudt3 is active on polyP when Zn2+ is present. Nudt3 has in vivo polyP phosphatase activity in human cells, and importantly, we show that cells with altered polyP levels by modifying Nudt3 protein amount present reduced viability upon oxidative stress and increased DNA damage, suggesting that polyP and Nudt3 play a role in oxidative stress protection. Finally, we show that Nudt3 is involved in the early stages of embryo development in zebrafish.
Collapse
Affiliation(s)
- Bàrbara Samper-Martín
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Ana Sarrias
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Blanca Lázaro
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Marta Pérez-Montero
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Rosalía Rodríguez-Rodríguez
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Mariana P C Ribeiro
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Aitor Bañón
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica, 08003 Barcelona, Spain
| | - Don Wolfgeher
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Henning J Jessen
- Institute of Organic Chemistry, University of Freiburg, 79104 Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Berta Alsina
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra-Parc de Recerca Biomèdica, 08003 Barcelona, Spain
| | - Josep Clotet
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain
| | - Stephen J Kron
- Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Adolfo Saiardi
- Medical Research Council Laboratory for Molecular Cell Biology, University College London, London WC1E6BT, UK
| | - Javier Jiménez
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain.
| | - Samuel Bru
- Faculty of Medicine and Health Sciences, Universitat Internacional de Catalunya, 08195 Sant Cugat del Vallès, Barcelona, Spain; Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Cerdanyola del Vallès, Spain.
| |
Collapse
|
160
|
Quick RE, Buck LD, Parab S, Tolbert ZR, Matsuoka RL. Highly Efficient Synthetic CRISPR RNA/Cas9-Based Mutagenesis for Rapid Cardiovascular Phenotypic Screening in F0 Zebrafish. Front Cell Dev Biol 2021; 9:735598. [PMID: 34746131 PMCID: PMC8570140 DOI: 10.3389/fcell.2021.735598] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/01/2021] [Indexed: 12/13/2022] Open
Abstract
The zebrafish is a valuable vertebrate model to study cardiovascular formation and function due to the facile visualization and rapid development of the circulatory system in its externally growing embryos. Despite having distinct advantages, zebrafish have paralogs of many important genes, making reverse genetics approaches inefficient since generating animals bearing multiple gene mutations requires substantial efforts. Here, we present a simple and robust synthetic CRISPR RNA/Cas9-based mutagenesis approach for generating biallelic F0 zebrafish knockouts. Using a dual-guide synthetic CRISPR RNA/Cas9 ribonucleoprotein (dgRNP) system, we compared the efficiency of biallelic gene disruptions following the injections of one, two, and three dgRNPs per gene into the cytoplasm or yolk. We show that simultaneous cytoplasmic injections of three distinct dgRNPs per gene into one-cell stage embryos resulted in the most efficient and consistent biallelic gene disruptions. Importantly, this triple dgRNP approach enables efficient inactivation of cell autonomous and cell non-autonomous gene function, likely due to the low mosaicism of biallelic disruptions. In support of this finding, we provide evidence that the F0 animals generated by this method fully phenocopied the endothelial and peri-vascular defects observed in corresponding stable mutant homozygotes. Moreover, this approach faithfully recapitulated the trunk vessel phenotypes resulting from the genetic interaction between two vegfr2 zebrafish paralogs. Mechanistically, investigation of genome editing and mRNA decay indicates that the combined mutagenic actions of three dgRNPs per gene lead to an increased probability of frameshift mutations, enabling efficient biallelic gene disruptions. Therefore, our approach offers a highly robust genetic platform to quickly assess novel and redundant gene function in F0 zebrafish.
Collapse
Affiliation(s)
- Rachael E Quick
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Luke D Buck
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Sweta Parab
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Zane R Tolbert
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Ryota L Matsuoka
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH, United States.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
161
|
Pant DC, Nazarko TY. Selective autophagy: the rise of the zebrafish model. Autophagy 2021; 17:3297-3305. [PMID: 33228439 PMCID: PMC8632090 DOI: 10.1080/15548627.2020.1853382] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022] Open
Abstract
Selective autophagy is a specific elimination of certain intracellular substrates by autophagic pathways. The most studied macroautophagy pathway involves tagging and recognition of a specific cargo by the autophagic membrane (phagophore) followed by the complete sequestration of targeted cargo from the cytosol by the double-membrane vesicle, autophagosome. Until recently, the knowledge about selective macroautophagy was minimal, but now there is a panoply of links elucidating how phagophores engulf their substrates selectively. The studies of selective autophagy processes have further stressed the importance of using the in vivo models to validate new in vitro findings and discover the physiologically relevant mechanisms. However, dissecting how the selective autophagy occurs yet remains difficult in living organisms, because most of the organelles are relatively inaccessible to observation and experimental manipulation in mammals. In recent years, zebrafish (Danio rerio) is widely recognized as an excellent model for studying autophagic processes in vivo because of its optical accessibility, genetic manipulability and translational potential. Several selective autophagy pathways, such as mitophagy, xenophagy, lipophagy and aggrephagy, have been investigated using zebrafish and still need to be studied further, while other selective autophagy pathways, such as pexophagy or reticulophagy, could also benefit from the use of the zebrafish model. In this review, we shed light on how zebrafish contributed to our understanding of these selective autophagy processes by providing the in vivo platform to study them at the organismal level and highlighted the versatility of zebrafish model in the selective autophagy field.Abbreviations: AD: Alzheimer disease; ALS: amyotrophic lateral sclerosis; Atg: autophagy-related; CMA: chaperone-mediated autophagy; CQ: chloroquine; HsAMBRA1: human AMBRA1; KD: knockdown; KO: knockout; LD: lipid droplet; MMA: methylmalonic acidemia; PD: Parkinson disease; Tg: transgenic.
Collapse
Affiliation(s)
- Devesh C. Pant
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Taras Y. Nazarko
- Department of Biology, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
162
|
Derrick CJ, Pollitt EJG, Sanchez Sevilla Uruchurtu A, Hussein F, Grierson AJ, Noël ES. Lamb1a regulates atrial growth by limiting second heart field addition during zebrafish heart development. Development 2021; 148:272294. [PMID: 34568948 DOI: 10.1242/dev.199691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
During early vertebrate heart development, the heart transitions from a linear tube to a complex asymmetric structure, a morphogenetic process that occurs simultaneously with growth of the heart. Cardiac growth during early heart morphogenesis is driven by deployment of cells from the second heart field (SHF) into both poles of the heart. Laminin is a core component of the extracellular matrix and, although mutations in laminin subunits are linked with cardiac abnormalities, no role for laminin has been identified in early vertebrate heart morphogenesis. We identified tissue-specific expression of laminin genes in the developing zebrafish heart, supporting a role for laminins in heart morphogenesis. Analysis of heart development in lamb1a zebrafish mutant embryos reveals mild morphogenetic defects and progressive cardiomegaly, and that Lamb1a functions to limit heart size during cardiac development by restricting SHF addition. lamb1a mutants exhibit hallmarks of altered haemodynamics, and blocking cardiac contractility in lamb1a mutants rescues heart size and atrial SHF addition. Together, these results suggest that laminin mediates interactions between SHF deployment and cardiac biomechanics during heart morphogenesis and growth in the developing embryo.
Collapse
Affiliation(s)
| | - Eric J G Pollitt
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | | | - Farah Hussein
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Andrew J Grierson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Emily S Noël
- Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
163
|
Moravec CE, Voit GC, Otterlee J, Pelegri F. Identification of maternal-effect genes in zebrafish using maternal crispants. Development 2021; 148:dev199536. [PMID: 34463742 PMCID: PMC8543149 DOI: 10.1242/dev.199536] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 08/19/2021] [Indexed: 01/25/2023]
Abstract
In animals, early development is dependent on a pool of maternal factors, both RNA and proteins, which are required for basic cellular processes and cell differentiation until zygotic genome activation. The role of the majority of these maternally expressed factors is not fully understood. By exploiting the biallelic editing ability of CRISPR-Cas9, we identify and characterize maternal-effect genes in a single generation, using a maternal crispant technique. We validated the ability to generate biallelic mutations in the germ line by creating maternal crispants that phenocopied previously characterized maternal-effect genes: birc5b, tmi and mid1ip1. Additionally, by targeting maternally expressed genes of unknown function in zebrafish, we identified two maternal-effect zebrafish genes, kpna7 and fhdc3. The genetic identity of these maternal crispants was confirmed by sequencing haploid progeny from F0 females, which allowed the analysis of newly induced lesions in the maternal germ line. Our studies show that maternal crispants allow for the effective identification and primary characterization of maternal-effect genes in a single generation, facilitating the reverse genetics analysis of maternal factors that drive embryonic development.
Collapse
Affiliation(s)
| | | | | | - Francisco Pelegri
- Laboratory of Genetics, University of Wisconsin-Madison, WI 53706, USA
| |
Collapse
|
164
|
Ttc30a affects tubulin modifications in a model for ciliary chondrodysplasia with polycystic kidney disease. Proc Natl Acad Sci U S A 2021; 118:2106770118. [PMID: 34548398 PMCID: PMC8488674 DOI: 10.1073/pnas.2106770118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
Cilia are tubulin-based cellular appendages, and their dysfunction has been linked to a variety of genetic diseases. Ciliary chondrodysplasia is one such condition that can co-occur with cystic kidney disease and other organ manifestations. We modeled skeletal ciliopathies by mutating two established disease genes in Xenopus tropicalis frogs. Bioinformatic analysis identified ttc30a as a ciliopathy network component, and targeting it replicated skeletal malformations and renal cysts as seen in patients and the amphibian models. A loss of Ttc30a affected cilia by altering posttranslational tubulin modifications. Our findings identify TTC30A/B as a component of ciliary segmentation essential for cartilage differentiation and renal tubulogenesis. These findings may lead to novel therapeutic targets in treating ciliary skeletopathies and cystic kidney disease. Skeletal ciliopathies (e.g., Jeune syndrome, short rib polydactyly syndrome, and Sensenbrenner syndrome) are frequently associated with nephronophthisis-like cystic kidney disease and other organ manifestations. Despite recent progress in genetic mapping of causative loci, a common molecular mechanism of cartilage defects and cystic kidneys has remained elusive. Targeting two ciliary chondrodysplasia loci (ift80 and ift172) by CRISPR/Cas9 mutagenesis, we established models for skeletal ciliopathies in Xenopus tropicalis. Froglets exhibited severe limb deformities, polydactyly, and cystic kidneys, closely matching the phenotype of affected patients. A data mining–based in silico screen found ttc30a to be related to known skeletal ciliopathy genes. CRISPR/Cas9 targeting replicated limb malformations and renal cysts identical to the models of established disease genes. Loss of Ttc30a impaired embryonic renal excretion and ciliogenesis because of altered posttranslational tubulin acetylation, glycylation, and defective axoneme compartmentalization. Ttc30a/b transcripts are enriched in chondrocytes and osteocytes of single-cell RNA-sequenced embryonic mouse limbs. We identify TTC30A/B as an essential node in the network of ciliary chondrodysplasia and nephronophthisis-like disease proteins and suggest that tubulin modifications and cilia segmentation contribute to skeletal and renal ciliopathy manifestations of ciliopathies in a cell type–specific manner. These findings have implications for potential therapeutic strategies.
Collapse
|
165
|
Zou YL, Ye AJ, Liu S, Wu WT, Xu LF, Dai FY, Tong XL. Expansion of targetable sites for the ribonucleoprotein-based CRISPR/Cas9 system in the silkworm Bombyx mori. BMC Biotechnol 2021; 21:54. [PMID: 34544395 PMCID: PMC8454041 DOI: 10.1186/s12896-021-00714-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/08/2021] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND With the emergence of CRISPR/Cas9 technology, multiple gene editing procedures became available for the silkworm. Although binary transgene-based methods have been widely used to generate mutants, delivery of the CRISPR/Cas9 system via DNA-free ribonucleoproteins offers several advantages. However, the T7 promoter that is widely used in the ribonucleoprotein-based method for production of sgRNAs in vitro requires a 5' GG motif for efficient initiation. The resulting transcripts bear a 5' GG motif, which significantly constrains the number of targetable sites in the silkworm genome. RESULTS In this study, we used the T7 promoter to add two supernumerary G residues to the 5' end of conventional (perfectly matched) 20-nucleotide sgRNA targeting sequences. We then asked if sgRNAs with this structure can generate mutations even if the genomic target does not contain corresponding GG residues. As expected, 5' GG mismatches depress the mutagenic activity of sgRNAs, and a single 5' G mismatch has a relatively minor effect. However, tests involving six sgRNAs targeting two genes show that the mismatches do not eliminate mutagenesis in vivo, and the efficiencies remain at useable levels. One sgRNA with a 5' GG mismatch at its target performed mutagenesis more efficiently than a conventional sgRNA with 5' matched GG residues at a second target within the same gene. Mutations generated by sgRNAs with 5' GG mismatches are also heritable. We successfully obtained null mutants with detectable phenotypes from sib-mated mosaics after one generation. CONCLUSIONS In summary, our method improves the utility and flexibility of the ribonucleoprotein-based CRISPR/Cas9 system in silkworm.
Collapse
Affiliation(s)
- Yun-Long Zou
- State Key Laboratory of Silkworm Genome Biology; Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs; College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Ai-Jun Ye
- State Key Laboratory of Silkworm Genome Biology; Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs; College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Shuo Liu
- State Key Laboratory of Silkworm Genome Biology; Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs; College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Wen-Tao Wu
- State Key Laboratory of Silkworm Genome Biology; Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs; College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Li-Feng Xu
- State Key Laboratory of Silkworm Genome Biology; Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs; College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Fang-Yin Dai
- State Key Laboratory of Silkworm Genome Biology; Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs; College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, People's Republic of China
| | - Xiao-Ling Tong
- State Key Laboratory of Silkworm Genome Biology; Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs; College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, People's Republic of China.
| |
Collapse
|
166
|
Luo K, Stocker R, Britton WJ, Kikuchi K, Oehlers SH. Haem oxygenase limits Mycobacterium marinum infection-induced detrimental ferrostatin-sensitive cell death in zebrafish. FEBS J 2021; 289:671-681. [PMID: 34544203 DOI: 10.1111/febs.16209] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 12/14/2022]
Abstract
Iron homeostasis is essential for both sides of the host-pathogen interface. Restricting access of iron slows bacterial growth while iron is also a necessary cofactor for host immunity. Haem oxygenase 1 (HMOX1) is a critical regulator of iron homeostasis that catalyses the liberation of iron during degradation of haem. It is also a stress-responsive protein that can be rapidly upregulated and confers protection to the host. Although a protective role of HMOX1 has been demonstrated in a variety of diseases, the role of HMOX1 in Mycobacterium tuberculosis infection is equivocal across experiments with different host-pathogen combinations. Here, we use the natural host-pathogen pairing of the zebrafish-Mycobacterium marinum infection platform to study the role of zebrafish haem oxygenase in mycobacterial infection. We identify zebrafish Hmox1a as the relevant functional paralog of mammalian HMOX1 and demonstrate a conserved role for Hmox1a in protecting the host from M. marinum infection. Using genetic and chemical tools, we show zebrafish Hmox1a protects the host against M. marinum infection by reducing infection-induced iron accumulation and ferrostatin-sensitive cell death.
Collapse
Affiliation(s)
- Kaiming Luo
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Roland Stocker
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,The Heart Research Institute, Newtown, NSW, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.,Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Kazu Kikuchi
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia.,Infection, Immunity and Inflammation Theme and Sydney Institute for Infectious Diseases, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
167
|
Parvez S, Herdman C, Beerens M, Chakraborti K, Harmer ZP, Yeh JRJ, MacRae CA, Yost HJ, Peterson RT. MIC-Drop: A platform for large-scale in vivo CRISPR screens. Science 2021; 373:1146-1151. [PMID: 34413171 DOI: 10.1126/science.abi8870] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Saba Parvez
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Chelsea Herdman
- Department of Neurobiology and Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Manu Beerens
- Department of Cardiovascular Medicine, Genetics and Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Korak Chakraborti
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Zachary P Harmer
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| | - Jing-Ruey J Yeh
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Calum A MacRae
- Department of Cardiovascular Medicine, Genetics and Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - H Joseph Yost
- Department of Neurobiology and Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Randall T Peterson
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
168
|
Presnell JS, Browne WE. Krüppel-like factor gene function in the ctenophore Mnemiopsis leidyi assessed by CRISPR/Cas9-mediated genome editing. Development 2021; 148:272041. [PMID: 34373891 DOI: 10.1242/dev.199771] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 07/26/2021] [Indexed: 12/21/2022]
Abstract
The Krüppel-like factor (Klf) gene family encodes transcription factors that play an important role in the regulation of stem cell proliferation, cell differentiation and development in bilaterians. Although Klf genes have been shown to specify functionally various cell types in non-bilaterian animals, their role in early-diverging animal lineages has not been assessed. Thus, the ancestral activity of these transcription factors in animal development is not well understood. The ctenophore Mnemiopsis leidyi has emerged as an important non-bilaterian model system for understanding early animal evolution. Here, we characterize the expression and functional role of Klf genes during M. leidyi embryogenesis. Zygotic Klf gene function was assessed with both CRISPR/Cas9-mediated genome editing and splice-blocking morpholino oligonucleotide knockdown approaches. Abrogation of zygotic Klf expression during M. leidyi embryogenesis resulted in abnormal development of several organs, including the pharynx, tentacle bulbs and apical organ. Our data suggest an ancient role for Klf genes in regulating endodermal patterning, possibly through regulation of cell proliferation.
Collapse
Affiliation(s)
- Jason S Presnell
- Department of Biology, University of Miami, Cox Science Center, 1301 Memorial Drive, Miami, FL 33146, USA
| | - William E Browne
- Department of Biology, University of Miami, Cox Science Center, 1301 Memorial Drive, Miami, FL 33146, USA
| |
Collapse
|
169
|
Progress in Gene-Editing Technology of Zebrafish. Biomolecules 2021; 11:biom11091300. [PMID: 34572513 PMCID: PMC8468279 DOI: 10.3390/biom11091300] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 08/25/2021] [Accepted: 08/30/2021] [Indexed: 12/26/2022] Open
Abstract
As a vertebrate model, zebrafish (Danio rerio) plays a vital role in the field of life sciences. Recently, gene-editing technology has become increasingly innovative, significantly promoting scientific research on zebrafish. However, the implementation of these methods in a reasonable and accurate manner to achieve efficient gene-editing remains challenging. In this review, we systematically summarize the development and latest progress in zebrafish gene-editing technology. Specifically, we outline trends in double-strand break-free genome modification and the prospective applications of fixed-point orientation transformation of any base at any location through a multi-method approach.
Collapse
|
170
|
Bek JW, Shochat C, De Clercq A, De Saffel H, Boel A, Metz J, Rodenburg F, Karasik D, Willaert A, Coucke PJ. Lrp5 Mutant and Crispant Zebrafish Faithfully Model Human Osteoporosis, Establishing the Zebrafish as a Platform for CRISPR-Based Functional Screening of Osteoporosis Candidate Genes. J Bone Miner Res 2021; 36:1749-1764. [PMID: 33957005 DOI: 10.1002/jbmr.4327] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 04/22/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
Genomewide association studies (GWAS) have improved our understanding of the genetic architecture of common complex diseases such as osteoporosis. Nevertheless, to attribute functional skeletal contributions of candidate genes to osteoporosis-related traits, there is a need for efficient and cost-effective in vivo functional testing. This can be achieved through CRISPR-based reverse genetic screens, where phenotyping is traditionally performed in stable germline knockout (KO) mutants. Recently it was shown that first-generation (F0) mosaic mutant zebrafish (so-called crispants) recapitulate the phenotype of germline KOs. To demonstrate feasibility of functional validation of osteoporosis candidate genes through crispant screening, we compared a crispant to a stable KO zebrafish model for the lrp5 gene. In humans, recessive loss-of-function mutations in LRP5, a co-receptor in the Wnt signaling pathway, cause osteoporosis-pseudoglioma syndrome. In addition, several GWAS studies identified LRP5 as a major risk locus for osteoporosis-related phenotypes. In this study, we showed that early stage lrp5 KO larvae display decreased notochord mineralization and malformations of the head cartilage. Quantitative micro-computed tomography (micro-CT) scanning and mass-spectrometry element analysis of the adult skeleton revealed decreased vertebral bone volume and bone mineralization, hallmark features of osteoporosis. Furthermore, regenerating fin tissue displayed reduced Wnt signaling activity in lrp5 KO adults. We next compared lrp5 mutants with crispants. Next-generation sequencing analysis of adult crispant tissue revealed a mean out-of-frame mutation rate of 76%, resulting in strongly reduced levels of Lrp5 protein. These crispants generally showed a milder but nonetheless highly comparable skeletal phenotype and a similarly reduced Wnt pathway response compared with lrp5 KO mutants. In conclusion, we show through faithful modeling of LRP5-related primary osteoporosis that crispant screening in zebrafish is a promising approach for rapid functional screening of osteoporosis candidate genes. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Jan Willem Bek
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Chen Shochat
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Adelbert De Clercq
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Hanna De Saffel
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Annekatrien Boel
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Department for Reproductive Medicine, Ghent University-University Hospital, Ghent, Belgium
| | - Juriaan Metz
- Department of Animal Ecology and Physiology, Radboud University, Nijmegen, The Netherlands
| | - Frans Rodenburg
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo, Japan.,Institute of Biology, Leiden University, Leiden, The Netherlands.,Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - David Karasik
- The Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.,Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, Boston, MA, USA
| | - Andy Willaert
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Paul J Coucke
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
171
|
Zhang C, Li J, Tarique I, Zhang Y, Lu T, Wang J, Chen A, Wen F, Zhang Z, Zhang Y, Shao M. A Time-Saving Strategy to Generate Double Maternal Mutants by an Oocyte-Specific Conditional Knockout System in Zebrafish. BIOLOGY 2021; 10:biology10080777. [PMID: 34440009 PMCID: PMC8389640 DOI: 10.3390/biology10080777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/29/2021] [Accepted: 08/14/2021] [Indexed: 12/23/2022]
Abstract
Simple Summary Maternally supplied mRNAs and proteins, termed maternal factors, are produced by over 14,000 coding genes in zebrafish. They play exclusive roles in controlling the formation of oocytes and the development of early embryos. These maternal factors can also compensate for the loss of function of its corresponding zygotic gene products. Thus, eliminating both maternal and zygotic gene products is essential to elucidate the functions of more than half of zebrafish genes. However, it is always challenging to inactivate maternal factors, because traditional genetic methods are either technically demanding or time-consuming. Our recent work established a rapid conditional knockout method to generate maternal or maternal and zygotic mutants in one fish generation. Here, we further test the feasibility of this approach to knock out two maternal genes with functional redundancy simultaneously. As a proof of principle, we successfully generated double maternal mutant embryos for dvl2 and dvl3a genes in three months for the first time. The cell movement defects in mutant embryos obtained by this approach mimic the genuine mutant embryos generated after fifteen months of time-consuming screening following the previously reported mosaic strategy. Therefore, this method has the potential to speed up the functional study of paralogous maternal genes. Abstract Maternal products are those mRNAs and proteins deposited during oogenesis, which play critical roles in controlling oocyte formation, fertilization, and early embryonic development. However, loss-of-function studies for these maternal factors are still lacking, mainly because of the prolonged period of transgenerational screening and technical barriers that prevent the generation of maternal (M) and maternal and zygotic (MZ) mutant embryos. By the transgenic expression of multiple sgRNAs targeting a single gene of interest in the background of a transgenic line Tg(zpc:zcas9) with oocyte-specific cas9 expression, we have successfully obtained maternal or maternal–zygotic mutant for single genes in F1 embryos. In this work, we tandemly connected a maternal GFP marker and eight sgRNA expression units to target dvl2 and dvl3a simultaneously and introduced this construct to the genome of Tg(zpc:zcas9) by meganuclease I-Sce I. As expected, we confirmed the existence of Mdvl2;Mdvl3a embryos with strong defective convergence and extension movement during gastrulation among outcrossed GFP positive F1 offspring. The MZdvl2;MZdvl3a embryos were also obtained by crossing the mutant carrying mosaic F0 female with dvl2+/−;dvl3a−/− male fish. This proof-of-principle thus highlights the potential of this conditional knockout strategy to circumvent the current difficulty in the study of genes with multiple functionally redundant paralogs.
Collapse
Affiliation(s)
- Chong Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Jiaguang Li
- Taishan College, Shandong University, Qingdao 266237, China; (J.L.); (Z.Z.)
| | - Imran Tarique
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Yizhuang Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Tong Lu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Jiasheng Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Aijun Chen
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Fenfen Wen
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Zhuoyu Zhang
- Taishan College, Shandong University, Qingdao 266237, China; (J.L.); (Z.Z.)
| | - Yanjun Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Ming Shao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
- Taishan College, Shandong University, Qingdao 266237, China; (J.L.); (Z.Z.)
- Correspondence:
| |
Collapse
|
172
|
Lu J, Fang W, Huang J, Li S. The application of genome editing technology in fish. MARINE LIFE SCIENCE & TECHNOLOGY 2021; 3:326-346. [PMID: 37073287 PMCID: PMC10077250 DOI: 10.1007/s42995-021-00091-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 01/11/2021] [Indexed: 05/03/2023]
Abstract
The advent and development of genome editing technology has opened up the possibility of directly targeting and modifying genomic sequences in the field of life sciences with rapid developments occurring in the last decade. As a powerful tool to decipher genome data at the molecular biology level, genome editing technology has made important contributions to elucidating many biological problems. Currently, the three most widely used genome editing technologies include: zinc finger nucleases (ZFN), transcription activator like effector nucleases (TALEN), and clustered regularly interspaced short palindromic repeats (CRISPR). Researchers are still striving to create simpler, more efficient, and accurate techniques, such as engineered base editors and new CRISPR/Cas systems, to improve editing efficiency and reduce off-target rate, as well as a near-PAMless SpCas9 variants to expand the scope of genome editing. As one of the important animal protein sources, fish has significant economic value in aquaculture. In addition, fish is indispensable for research as it serves as the evolutionary link between invertebrates and higher vertebrates. Consequently, genome editing technologies were applied extensively in various fish species for basic functional studies as well as applied research in aquaculture. In this review, we focus on the application of genome editing technologies in fish species detailing growth, gender, and pigmentation traits. In addition, we have focused on the construction of a zebrafish (Danio rerio) disease model and high-throughput screening of functional genes. Finally, we provide some of the future perspectives of this technology.
Collapse
Affiliation(s)
- Jianguo Lu
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai, 519082 China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519080 China
| | - Wenyu Fang
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai, 519082 China
| | - Junrou Huang
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai, 519082 China
| | - Shizhu Li
- School of Marine Sciences, Sun Yat-Sen University, Zhuhai, 519082 China
| |
Collapse
|
173
|
Zhang C, Lu T, Zhang Y, Li J, Tarique I, Wen F, Chen A, Wang J, Zhang Z, Zhang Y, Shi DL, Shao M. Rapid generation of maternal mutants via oocyte transgenic expression of CRISPR-Cas9 and sgRNAs in zebrafish. SCIENCE ADVANCES 2021; 7:eabg4243. [PMID: 34362733 PMCID: PMC8346210 DOI: 10.1126/sciadv.abg4243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/21/2021] [Indexed: 05/08/2023]
Abstract
Maternal products are exclusive factors to drive oogenesis and early embryonic development. As disrupting maternal gene functions is either time-consuming or technically challenging, early developmental programs regulated by maternal factors remain mostly elusive. We provide a transgenic approach to inactivate maternal genes in zebrafish primary oocytes. By introducing three tandem single guide RNA (sgRNA) expression cassettes and a green fluorescent protein (GFP) reporter into Tg(zpc:zcas9) embryos, we efficiently obtained maternal nanog and ctnnb2 mutants among GFP-positive F1 offspring. Notably, most of these maternal mutants displayed either sgRNA site-spanning genomic deletions or unintended large deletions extending distantly from the sgRNA targets, suggesting a prominent deletion-prone tendency of genome editing in the oocyte. Thus, our method allows maternal gene knockout in the absence of viable and fertile homozygous mutant adults. This approach is particularly time-saving and can be applied for functional screening of maternal factors and generating genomic deletions in zebrafish.
Collapse
Affiliation(s)
- Chong Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Tong Lu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Yizhuang Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Jiaguang Li
- Shandong University Taishan College, Qingdao 266237, China
| | - Imran Tarique
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Fenfen Wen
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Aijun Chen
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Jiasheng Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Zhuoyu Zhang
- Shandong University Taishan College, Qingdao 266237, China
| | - Yanjun Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - De-Li Shi
- Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
- Developmental Biology Laboratory, CNRS-UMR7622, Institut de Biologie Paris-Seine, Sorbonne University, Paris 75005, France
| | - Ming Shao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China.
- Shandong University Taishan College, Qingdao 266237, China
| |
Collapse
|
174
|
Marchal GA, Jouni M, Chiang DY, Pérez-Hernández M, Podliesna S, Yu N, Casini S, Potet F, Veerman CC, Klerk M, Lodder EM, Mengarelli I, Guan K, Vanoye CG, Rothenberg E, Charpentier F, Redon R, George AL, Verkerk AO, Bezzina CR, MacRae CA, Burridge PW, Delmar M, Galjart N, Portero V, Remme CA. Targeting the Microtubule EB1-CLASP2 Complex Modulates Na V1.5 at Intercalated Discs. Circ Res 2021; 129:349-365. [PMID: 34092082 PMCID: PMC8298292 DOI: 10.1161/circresaha.120.318643] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mariam Jouni
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - David Y Chiang
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | | | - Svitlana Podliesna
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Nuo Yu
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Simona Casini
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Franck Potet
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Christiaan C Veerman
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Elisabeth M Lodder
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Isabella Mengarelli
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Kaomei Guan
- Institute of Pharmacology and Toxicology, Technische Universität Dresden, Germany (K.G.)
| | - Carlos G Vanoye
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Eli Rothenberg
- Department of Biochemistry and Pharmacology (E.R.), NYU School of Medicine
| | - Flavien Charpentier
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Richard Redon
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Alfred L George
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Arie O Verkerk
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Department of Medical Biology, Amsterdam UMC - location AMC, The Netherlands (M.K., A.O.V.)
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| | - Calum A MacRae
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA (D.Y.C., C.A.M.)
| | - Paul W Burridge
- Department of Pharmacology, University Feinberg School of Medicine, Chicago, IL (M.J., F.P., C.G.V., A.L.G., P.W.B.)
| | - Mario Delmar
- Division of Cardiology (M.P.-H., M.D.), NYU School of Medicine
| | - Niels Galjart
- Department of Cell Biology, Erasmus Medical Centre Rotterdam, The Netherlands (N.Y., N.G.)
| | - Vincent Portero
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
- Université de Nantes, CNRS, INSERM, l'institut du Thorax, Nantes, France (F.C., R.R., V.P.)
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC - location AMC, The Netherlands (G.A.M., S.P., S.C., C.C.V., E.M.L., I.M., A.O.V., C.R.B., V.P., C.A.R.)
| |
Collapse
|
175
|
Blitz IL, Nakayama T. CRISPR-Cas9 Mutagenesis in Xenopus tropicalis for Phenotypic Analyses in the F 0 Generation and Beyond. Cold Spring Harb Protoc 2021; 2022:pdb.prot106971. [PMID: 34244352 DOI: 10.1101/pdb.prot106971] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
CRISPR-Cas9 mutagenesis is being widely used to create targeted loss-of-function mutations in the diploid frog Xenopus tropicalis Here we describe a simple mutagenesis protocol using microinjection of Cas9 protein or mRNA, together with synthetic guide RNAs (sgRNAs) targeting specific DNA sequences, into the early embryo. Cas9-catalyzed double-strand breaks undergo error-prone repair, resulting in production of short insertions and/or deletions. Thus, careful selection of target sites can lead to mutations that impair normal function of the protein product. CRISPR-Cas9 can be used to create either mosaic loss-of-function Xenopus embryos that display F0 generation phenotypes or mutant lines for downstream analysis. In addition to describing how to mutagenize genes using CRISPR-Cas9, we also discuss a simple method to determine the mutagenesis efficiency, some potential problems that can arise, and possible solutions to overcome them. The protocol described here should be applicable to other amphibians and, in principle, many other organisms.
Collapse
Affiliation(s)
- Ira L Blitz
- Department of Developmental and Cell Biology, University of California, Irvine, California 92697, USA
| | - Takuya Nakayama
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904, USA
| |
Collapse
|
176
|
Zhang L, Chen C, Fu J, Lilley B, Berlinicke C, Hansen B, Ding D, Wang G, Wang T, Shou D, Ye Y, Mulligan T, Emmerich K, Saxena MT, Hall KR, Sharrock AV, Brandon C, Park H, Kam TI, Dawson VL, Dawson TM, Shim JS, Hanes J, Ji H, Liu JO, Qian J, Ackerley DF, Rohrer B, Zack DJ, Mumm JS. Large-scale phenotypic drug screen identifies neuroprotectants in zebrafish and mouse models of retinitis pigmentosa. eLife 2021; 10:e57245. [PMID: 34184634 PMCID: PMC8425951 DOI: 10.7554/elife.57245] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 06/28/2021] [Indexed: 11/25/2022] Open
Abstract
Retinitis pigmentosa (RP) and associated inherited retinal diseases (IRDs) are caused by rod photoreceptor degeneration, necessitating therapeutics promoting rod photoreceptor survival. To address this, we tested compounds for neuroprotective effects in multiple zebrafish and mouse RP models, reasoning drugs effective across species and/or independent of disease mutation may translate better clinically. We first performed a large-scale phenotypic drug screen for compounds promoting rod cell survival in a larval zebrafish model of inducible RP. We tested 2934 compounds, mostly human-approved drugs, across six concentrations, resulting in 113 compounds being identified as hits. Secondary tests of 42 high-priority hits confirmed eleven lead candidates. Leads were then evaluated in a series of mouse RP models in an effort to identify compounds effective across species and RP models, that is, potential pan-disease therapeutics. Nine of 11 leads exhibited neuroprotective effects in mouse primary photoreceptor cultures, and three promoted photoreceptor survival in mouse rd1 retinal explants. Both shared and complementary mechanisms of action were implicated across leads. Shared target tests implicated parp1-dependent cell death in our zebrafish RP model. Complementation tests revealed enhanced and additive/synergistic neuroprotective effects of paired drug combinations in mouse photoreceptor cultures and zebrafish, respectively. These results highlight the value of cross-species/multi-model phenotypic drug discovery and suggest combinatorial drug therapies may provide enhanced therapeutic benefits for RP patients.
Collapse
Affiliation(s)
- Liyun Zhang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Conan Chen
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Jie Fu
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Brendan Lilley
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Cynthia Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Baranda Hansen
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Ding Ding
- Department of Biostatistics, Johns Hopkins UniversityBaltimoreUnited States
| | - Guohua Wang
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Tao Wang
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- School of Chemistry, Xuzhou College of Industrial TechnologyXuzhouChina
- College of Light Industry and Food Engineering, Nanjing Forestry UniversityNanjingChina
| | - Daniel Shou
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Ying Ye
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Timothy Mulligan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Kevin Emmerich
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- Department of Genetic Medicine, Johns Hopkins UniversityBaltimoreUnited States
| | - Meera T Saxena
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Kelsi R Hall
- School of Biological Sciences, Victoria University of WellingtonWellingtonNew Zealand
| | - Abigail V Sharrock
- Department of Biostatistics, Johns Hopkins UniversityBaltimoreUnited States
- School of Biological Sciences, Victoria University of WellingtonWellingtonNew Zealand
| | - Carlene Brandon
- Department of Ophthalmology, Medical University of South CarolinaCharlestonUnited States
| | - Hyejin Park
- Department of Neurology, Johns Hopkins UniversityBaltimoreUnited States
| | - Tae-In Kam
- Department of Neurology, Johns Hopkins UniversityBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins UniversityBaltimoreUnited States
| | - Valina L Dawson
- Department of Neurology, Johns Hopkins UniversityBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins UniversityBaltimoreUnited States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins UniversityBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| | - Ted M Dawson
- Department of Neurology, Johns Hopkins UniversityBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins UniversityBaltimoreUnited States
- Department of Pharmacology and Molecular Sciences, Johns Hopkins UniversityBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| | - Joong Sup Shim
- Faculty of Health Sciences, University of Macau, TaipaMacauChina
| | - Justin Hanes
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins UniversityBaltimoreUnited States
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins UniversityBaltimoreUnited States
- Department of Oncology, Johns Hopkins UniversityBaltimoreUnited States
| | - Jiang Qian
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
| | - David F Ackerley
- School of Biological Sciences, Victoria University of WellingtonWellingtonNew Zealand
| | - Baerbel Rohrer
- Department of Ophthalmology, Medical University of South CarolinaCharlestonUnited States
| | - Donald J Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- Department of Genetic Medicine, Johns Hopkins UniversityBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
- Department of Molecular Biology and Genetics, Johns Hopkins UniversityBaltimoreUnited States
| | - Jeff S Mumm
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- The Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins UniversityBaltimoreUnited States
- Department of Genetic Medicine, Johns Hopkins UniversityBaltimoreUnited States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins UniversityBaltimoreUnited States
| |
Collapse
|
177
|
Pioneer Axons Utilize a Dcc Signaling-Mediated Invasion Brake to Precisely Complete Their Pathfinding Odyssey. J Neurosci 2021; 41:6617-6636. [PMID: 34131031 DOI: 10.1523/jneurosci.0212-21.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/01/2021] [Accepted: 06/08/2021] [Indexed: 11/21/2022] Open
Abstract
Axons navigate through the embryo to construct a functional nervous system. A missing part of the axon navigation puzzle is how a single axon traverses distinct anatomic choice points through its navigation. The dorsal root ganglia (DRG) neurons experience such choice points. First, they navigate to the dorsal root entry zone (DREZ), then halt navigation in the peripheral nervous system to invade the spinal cord, and then reinitiate navigation inside the CNS. Here, we used time-lapse super-resolution imaging in zebrafish DRG pioneer neurons to investigate how embryonic axons control their cytoskeleton to navigate to and invade at the correct anatomic position. We found that invadopodia components form in the growth cone even during filopodia-based navigation, but only stabilize when the axon is at the spinal cord entry location. Further, we show that intermediate levels of DCC and cAMP, as well as Rac1 activation, subsequently engage an axon invasion brake. Our results indicate that actin-based invadopodia components form in the growth cone and disruption of the invasion brake causes axon entry defects and results in failed behavioral responses, thereby demonstrating the importance of regulating distinct actin populations during navigational challenges.SIGNIFICANCE STATEMENT Correct spatiotemporal navigation of neuronal growth cones is dependent on extracellular navigational cues and growth cone dynamics. Here, we link dcc-mediated signaling to actin-based invadopodia and filopodia dynamics during pathfinding and entry into the spinal cord using an in vivo model of dorsal root ganglia (DRG) sensory axons. We reveal a molecularly-controlled brake on invadopodia stabilization until the sensory neuron growth cone is present at the dorsal root entry zone (DREZ), which is ultimately essential for growth cone entry into the spinal cord and behavioral response.
Collapse
|
178
|
Guarin M, Faelens R, Giusti A, De Croze N, Léonard M, Cabooter D, Annaert P, de Witte P, Ny A. Spatiotemporal imaging and pharmacokinetics of fluorescent compounds in zebrafish eleuthero-embryos after different routes of administration. Sci Rep 2021; 11:12229. [PMID: 34108572 PMCID: PMC8190279 DOI: 10.1038/s41598-021-91612-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Zebrafish (Danio rerio) is increasingly used to assess the pharmacological activity and toxicity of compounds. The spatiotemporal distribution of seven fluorescent alkyne compounds was examined during 48 h after immersion (10 µM) or microinjection (2 mg/kg) in the pericardial cavity (PC), intraperitoneally (IP) and yolk sac (IY) of 3 dpf zebrafish eleuthero-embryos. By modelling the fluorescence of whole-body contours present in fluorescence images, the main pharmacokinetic (PK) parameter values of the compounds were determined. It was demonstrated that especially in case of short incubations (1-3 h) immersion can result in limited intrabody exposure to compounds. In this case, PC and IP microinjections represent excellent alternatives. Significantly, IY microinjections did not result in a suitable intrabody distribution of the compounds. Performing a QSPkR (quantitative structure-pharmacokinetic relationship) analysis, LogD was identified as the only molecular descriptor that explains the final uptake of the selected compounds. It was also shown that combined administration of compounds (immersion and microinjection) provides a more stable intrabody exposure, at least in case of a prolonged immersion and compounds with LogD value > 1. These results will help reduce the risk of false negative results and can offer an invaluable input for future translational research and safety assessment applications.
Collapse
Affiliation(s)
- Marlly Guarin
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Ruben Faelens
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Arianna Giusti
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | | | - Marc Léonard
- L'Oréal, Research and Innovation, Aulnay-sous-Bois, France
| | - Deirdre Cabooter
- Pharmaceutical Analysis, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Pieter Annaert
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium.
| | - Peter de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium.
| | - Annelii Ny
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
179
|
Sheets L, Holmgren M, Kindt KS. How Zebrafish Can Drive the Future of Genetic-based Hearing and Balance Research. J Assoc Res Otolaryngol 2021; 22:215-235. [PMID: 33909162 PMCID: PMC8110678 DOI: 10.1007/s10162-021-00798-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/23/2021] [Indexed: 02/06/2023] Open
Abstract
Over the last several decades, studies in humans and animal models have successfully identified numerous molecules required for hearing and balance. Many of these studies relied on unbiased forward genetic screens based on behavior or morphology to identify these molecules. Alongside forward genetic screens, reverse genetics has further driven the exploration of candidate molecules. This review provides an overview of the genetic studies that have established zebrafish as a genetic model for hearing and balance research. Further, we discuss how the unique advantages of zebrafish can be leveraged in future genetic studies. We explore strategies to design novel forward genetic screens based on morphological alterations using transgenic lines or behavioral changes following mechanical or acoustic damage. We also outline how recent advances in CRISPR-Cas9 can be applied to perform reverse genetic screens to validate large sequencing datasets. Overall, this review describes how future genetic studies in zebrafish can continue to advance our understanding of inherited and acquired hearing and balance disorders.
Collapse
Affiliation(s)
- Lavinia Sheets
- Department of Otolaryngology-Head & Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Melanie Holmgren
- Department of Otolaryngology-Head & Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Katie S Kindt
- Section On Sensory Cell Development and Function, National Institutes On Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, USA.
| |
Collapse
|
180
|
Lauri A, Fasano G, Venditti M, Dallapiccola B, Tartaglia M. In vivo Functional Genomics for Undiagnosed Patients: The Impact of Small GTPases Signaling Dysregulation at Pan-Embryo Developmental Scale. Front Cell Dev Biol 2021; 9:642235. [PMID: 34124035 PMCID: PMC8194860 DOI: 10.3389/fcell.2021.642235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
While individually rare, disorders affecting development collectively represent a substantial clinical, psychological, and socioeconomic burden to patients, families, and society. Insights into the molecular mechanisms underlying these disorders are required to speed up diagnosis, improve counseling, and optimize management toward targeted therapies. Genome sequencing is now unveiling previously unexplored genetic variations in undiagnosed patients, which require functional validation and mechanistic understanding, particularly when dealing with novel nosologic entities. Functional perturbations of key regulators acting on signals' intersections of evolutionarily conserved pathways in these pathological conditions hinder the fine balance between various developmental inputs governing morphogenesis and homeostasis. However, the distinct mechanisms by which these hubs orchestrate pathways to ensure the developmental coordinates are poorly understood. Integrative functional genomics implementing quantitative in vivo models of embryogenesis with subcellular precision in whole organisms contribute to answering these questions. Here, we review the current knowledge on genes and mechanisms critically involved in developmental syndromes and pediatric cancers, revealed by genomic sequencing and in vivo models such as insects, worms and fish. We focus on the monomeric GTPases of the RAS superfamily and their influence on crucial developmental signals and processes. We next discuss the effectiveness of exponentially growing functional assays employing tractable models to identify regulatory crossroads. Unprecedented sophistications are now possible in zebrafish, i.e., genome editing with single-nucleotide precision, nanoimaging, highly resolved recording of multiple small molecules activity, and simultaneous monitoring of brain circuits and complex behavioral response. These assets permit accurate real-time reporting of dynamic small GTPases-controlled processes in entire organisms, owning the potential to tackle rare disease mechanisms.
Collapse
Affiliation(s)
- Antonella Lauri
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | | | | | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| |
Collapse
|
181
|
Okuda KS, Keyser MS, Gurevich DB, Sturtzel C, Mason EA, Paterson S, Chen H, Scott M, Condon ND, Martin P, Distel M, Hogan BM. Live-imaging of endothelial Erk activity reveals dynamic and sequential signalling events during regenerative angiogenesis. eLife 2021; 10:62196. [PMID: 34003110 PMCID: PMC8175085 DOI: 10.7554/elife.62196] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 05/17/2021] [Indexed: 12/23/2022] Open
Abstract
The formation of new blood vessel networks occurs via angiogenesis during development, tissue repair, and disease. Angiogenesis is regulated by intracellular endothelial signalling pathways, induced downstream of vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs). A major challenge in understanding angiogenesis is interpreting how signalling events occur dynamically within endothelial cell populations during sprouting, proliferation, and migration. Extracellular signal-regulated kinase (Erk) is a central downstream effector of Vegf-signalling and reports the signalling that drives angiogenesis. We generated a vascular Erk biosensor transgenic line in zebrafish using a kinase translocation reporter that allows live-imaging of Erk-signalling dynamics. We demonstrate the utility of this line to live-image Erk activity during physiologically relevant angiogenic events. Further, we reveal dynamic and sequential endothelial cell Erk-signalling events following blood vessel wounding. Initial signalling is dependent upon Ca2+ in the earliest responding endothelial cells, but is independent of Vegfr-signalling and local inflammation. The sustained regenerative response, however, involves a Vegfr-dependent mechanism that initiates concomitantly with the wound inflammatory response. This work reveals a highly dynamic sequence of signalling events in regenerative angiogenesis and validates a new resource for the study of vascular Erk-signalling in real-time.
Collapse
Affiliation(s)
- Kazuhide S Okuda
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, St Lucia, Australia
| | - Mikaela S Keyser
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, St Lucia, Australia
| | - David B Gurevich
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, United Kingdom.,Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath, United Kingdom
| | - Caterina Sturtzel
- Innovative Cancer Models, St Anna Kinderkrebsforschung, Children's Cancer Research Institute, Vienna, Austria.,Zebrafish Platform Austria for preclinical drug screening (ZANDR), Vienna, Austria
| | - Elizabeth A Mason
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia
| | - Scott Paterson
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, St Lucia, Australia
| | - Huijun Chen
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, St Lucia, Australia
| | - Mark Scott
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, St Lucia, Australia
| | - Nicholas D Condon
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, St Lucia, Australia
| | - Paul Martin
- School of Biochemistry, Biomedical Sciences Building, University Walk, University of Bristol, Bristol, United Kingdom
| | - Martin Distel
- Innovative Cancer Models, St Anna Kinderkrebsforschung, Children's Cancer Research Institute, Vienna, Austria.,Zebrafish Platform Austria for preclinical drug screening (ZANDR), Vienna, Austria
| | - Benjamin M Hogan
- Organogenesis and Cancer Program, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Department of Anatomy and Physiology, University of Melbourne, Melbourne, Australia.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, St Lucia, Australia
| |
Collapse
|
182
|
Knickmeyer MD, Mateo JL, Heermann S. BMP Signaling Interferes with Optic Chiasm Formation and Retinal Ganglion Cell Pathfinding in Zebrafish. Int J Mol Sci 2021; 22:ijms22094560. [PMID: 33925390 PMCID: PMC8123821 DOI: 10.3390/ijms22094560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/19/2021] [Accepted: 04/24/2021] [Indexed: 11/17/2022] Open
Abstract
Decussation of axonal tracts is an important hallmark of vertebrate neuroanatomy resulting in one brain hemisphere controlling the contralateral side of the body and also computing the sensory information originating from that respective side. Here, we show that BMP interferes with optic chiasm formation and RGC pathfinding in zebrafish. Experimental induction of BMP4 at 15 hpf results in a complete ipsilateral projection of RGC axons and failure of commissural connections of the forebrain, in part as the result of an interaction with shh signaling, transcriptional regulation of midline guidance cues and an affected optic stalk morphogenesis. Experimental induction of BMP4 at 24 hpf, resulting in only a mild repression of forebrain shh ligand expression but in a broad expression of pax2a in the diencephalon, does not per se prevent RGC axons from crossing the midline. It nevertheless shows severe pathologies of RGC projections e.g., the fasciculation of RGC axons with the ipsilateral optic tract resulting in the innervation of one tectum by two eyes or the projection of RGC axons in the direction of the contralateral eye.
Collapse
Affiliation(s)
- Max D. Knickmeyer
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University Freiburg, 79104 Freiburg, Germany;
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, 79104 Freiburg, Germany
| | - Juan L. Mateo
- Departamento de Informática, Universidad de Oviedo, Jesús Arias de Velasco, 33005 Oviedo, Spain;
| | - Stephan Heermann
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Faculty of Medicine, University Freiburg, 79104 Freiburg, Germany;
- Correspondence:
| |
Collapse
|
183
|
Francis CR, Claflin S, Kushner EJ. Synaptotagmin-Like Protein 2a Regulates Angiogenic Lumen Formation via Weibel-Palade Body Apical Secretion of Angiopoietin-2. Arterioscler Thromb Vasc Biol 2021; 41:1972-1986. [PMID: 33853352 DOI: 10.1161/atvbaha.121.316113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
| | - Shea Claflin
- Department of Biological Sciences, University of Denver, CO
| | | |
Collapse
|
184
|
Keatinge M, Tsarouchas TM, Munir T, Porter NJ, Larraz J, Gianni D, Tsai HH, Becker CG, Lyons DA, Becker T. CRISPR gRNA phenotypic screening in zebrafish reveals pro-regenerative genes in spinal cord injury. PLoS Genet 2021; 17:e1009515. [PMID: 33914736 PMCID: PMC8084196 DOI: 10.1371/journal.pgen.1009515] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 03/28/2021] [Indexed: 12/30/2022] Open
Abstract
Zebrafish exhibit robust regeneration following spinal cord injury, promoted by macrophages that control post-injury inflammation. However, the mechanistic basis of how macrophages regulate regeneration is poorly understood. To address this gap in understanding, we conducted a rapid in vivo phenotypic screen for macrophage-related genes that promote regeneration after spinal injury. We used acute injection of synthetic RNA Oligo CRISPR guide RNAs (sCrRNAs) that were pre-screened for high activity in vivo. Pre-screening of over 350 sCrRNAs allowed us to rapidly identify highly active sCrRNAs (up to half, abbreviated as haCRs) and to effectively target 30 potentially macrophage-related genes. Disruption of 10 of these genes impaired axonal regeneration following spinal cord injury. We selected 5 genes for further analysis and generated stable mutants using haCRs. Four of these mutants (tgfb1a, tgfb3, tnfa, sparc) retained the acute haCR phenotype, validating the approach. Mechanistically, tgfb1a haCR-injected and stable mutant zebrafish fail to resolve post-injury inflammation, indicated by prolonged presence of neutrophils and increased levels of il1b expression. Inhibition of Il-1β rescues the impaired axon regeneration in the tgfb1a mutant. Hence, our rapid and scalable screening approach has identified functional regulators of spinal cord regeneration, but can be applied to any biological function of interest.
Collapse
Affiliation(s)
- Marcus Keatinge
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Tahimina Munir
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicola J. Porter
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Juan Larraz
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Davide Gianni
- Biogen, Cambridge, Massachusetts, United States of America
| | - Hui-Hsin Tsai
- Biogen, Cambridge, Massachusetts, United States of America
| | - Catherina G. Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David A. Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Thomas Becker
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
185
|
Wright K, de Silva K, Plain KM, Purdie AC, Blair TA, Duggin IG, Britton WJ, Oehlers SH. Mycobacterial infection-induced miR-206 inhibits protective neutrophil recruitment via the CXCL12/CXCR4 signalling axis. PLoS Pathog 2021; 17:e1009186. [PMID: 33826679 PMCID: PMC8055004 DOI: 10.1371/journal.ppat.1009186] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/19/2021] [Accepted: 03/29/2021] [Indexed: 12/22/2022] Open
Abstract
Pathogenic mycobacteria actively dysregulate protective host immune signalling pathways during infection to drive the formation of permissive granuloma microenvironments. Dynamic regulation of host microRNA (miRNA) expression is a conserved feature of mycobacterial infections across host-pathogen pairings. Here we examine the role of miR-206 in the zebrafish model of Mycobacterium marinum infection, which allows investigation of the early stages of granuloma formation. We find miR-206 is upregulated following infection by pathogenic M. marinum and that antagomir-mediated knockdown of miR-206 is protective against infection. We observed striking upregulation of cxcl12a and cxcr4b in infected miR-206 knockdown zebrafish embryos and live imaging revealed enhanced recruitment of neutrophils to sites of infection. We used CRISPR/Cas9-mediated knockdown of cxcl12a and cxcr4b expression and AMD3100 inhibition of Cxcr4 to show that the enhanced neutrophil response and reduced bacterial burden caused by miR-206 knockdown was dependent on the Cxcl12/Cxcr4 signalling axis. Together, our data illustrate a pathway through which pathogenic mycobacteria induce host miR-206 expression to suppress Cxcl12/Cxcr4 signalling and prevent protective neutrophil recruitment to granulomas.
Collapse
Affiliation(s)
- Kathryn Wright
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
- The University of Sydney, Faculty of Science, Sydney School of Veterinary Science, Sydney, New South Wales, Australia
| | - Kumudika de Silva
- The University of Sydney, Faculty of Science, Sydney School of Veterinary Science, Sydney, New South Wales, Australia
| | - Karren M. Plain
- The University of Sydney, Faculty of Science, Sydney School of Veterinary Science, Sydney, New South Wales, Australia
| | - Auriol C. Purdie
- The University of Sydney, Faculty of Science, Sydney School of Veterinary Science, Sydney, New South Wales, Australia
| | - Tamika A. Blair
- ithree Institute, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Iain G. Duggin
- ithree Institute, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Warwick J. Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
- Department of Clinical Immunology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Stefan H. Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, New South Wales, Australia
- The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, New South Wales, Australia
| |
Collapse
|
186
|
Bourgeois JS, Smith CM, Ko DC. These Are the Genes You're Looking For: Finding Host Resistance Genes. Trends Microbiol 2021; 29:346-362. [PMID: 33004258 PMCID: PMC7969353 DOI: 10.1016/j.tim.2020.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/21/2022]
Abstract
Humanity's ongoing struggle with new, re-emerging and endemic infectious diseases serves as a frequent reminder of the need to understand host-pathogen interactions. Recent advances in genomics have dramatically advanced our understanding of how genetics contributes to host resistance or susceptibility to bacterial infection. Here we discuss current trends in defining host-bacterial interactions at the genome-wide level, including screens that harness CRISPR/Cas9 genome editing, natural genetic variation, proteomics, and transcriptomics. We report on the merits, limitations, and findings of these innovative screens and discuss their complementary nature. Finally, we speculate on future innovation as we continue to progress through the postgenomic era and towards deeper mechanistic insight and clinical applications.
Collapse
Affiliation(s)
- Jeffrey S Bourgeois
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Clare M Smith
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA; Duke Human Vaccine Institute, School of Medicine, Duke University Durham, NC, USA
| | - Dennis C Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA; University Program in Genetics and Genomics, Duke University, Durham, NC, USA; Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
187
|
Li H, Yu H, Du S, Li Q. CRISPR/Cas9 Mediated High Efficiency Knockout of Myosin Essential Light Chain Gene in the Pacific Oyster (Crassostrea Gigas). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2021; 23:215-224. [PMID: 33715060 DOI: 10.1007/s10126-020-10016-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/27/2020] [Indexed: 06/12/2023]
Abstract
Pacific oyster (Crassostrea gigas) is one of the most widely cultivated shellfish species in the world. Because of its economic value and complex life cycle involving drastic changes from a free-swimming larva to a sessile juvenile, C. gigas has been used as a model for developmental, environmental, and aquaculture research. However, due to the lack of genetic tools for functional analysis, gene functions associated with biological or economic traits cannot be easily determined. Here, we reported a successful application of CRISPR/Cas9 technology for knockout of myosin essential light chain gene (CgMELC) in C. gigas. C. gigas embryos injected with sgRNAs/Cas9 contained extensive indel mutations at the target sites. The mutant larvae showed defective musculature and reduced motility. In addition, knockout of CgMELC disrupted the expression and patterning of myosin heavy chain positive myofibers in larvae. Together, these data indicate that CgMELC is involved in larval muscle contraction and myogenesis in oyster larvae.
Collapse
Affiliation(s)
- Huijuan Li
- Key Laboratory of Mariculture, (Ocean University of China), Ministry of Education, Qingdao, 266003, China
| | - Hong Yu
- Key Laboratory of Mariculture, (Ocean University of China), Ministry of Education, Qingdao, 266003, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shaojun Du
- Institute of Marine and Environmental Technology, Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Qi Li
- Key Laboratory of Mariculture, (Ocean University of China), Ministry of Education, Qingdao, 266003, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
188
|
Cronan MR, Hughes EJ, Brewer WJ, Viswanathan G, Hunt EG, Singh B, Mehra S, Oehlers SH, Gregory SG, Kaushal D, Tobin DM. A non-canonical type 2 immune response coordinates tuberculous granuloma formation and epithelialization. Cell 2021; 184:1757-1774.e14. [PMID: 33761328 PMCID: PMC8055144 DOI: 10.1016/j.cell.2021.02.046] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 11/03/2020] [Accepted: 02/22/2021] [Indexed: 12/19/2022]
Abstract
The central pathogen-immune interface in tuberculosis is the granuloma, a complex host immune structure that dictates infection trajectory and physiology. Granuloma macrophages undergo a dramatic transition in which entire epithelial modules are induced and define granuloma architecture. In tuberculosis, relatively little is known about the host signals that trigger this transition. Using the zebrafish-Mycobacterium marinum model, we identify the basis of granuloma macrophage transformation. Single-cell RNA-sequencing analysis of zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected macaques reveal that, even in the presence of robust type 1 immune responses, countervailing type 2 signals associate with macrophage epithelialization. We find that type 2 immune signaling, mediated via stat6, is absolutely required for epithelialization and granuloma formation. In mixed chimeras, stat6 acts cell autonomously within macrophages, where it is required for epithelioid transformation and incorporation into necrotic granulomas. These findings establish the signaling pathway that produces the hallmark structure of mycobacterial infection.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified/genetics
- Animals, Genetically Modified/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation
- Disease Models, Animal
- Epithelioid Cells/cytology
- Epithelioid Cells/immunology
- Epithelioid Cells/metabolism
- Granuloma/immunology
- Granuloma/metabolism
- Granuloma/pathology
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/metabolism
- Immunity/physiology
- Interferon-gamma/metabolism
- Interleukin-12/metabolism
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Mycobacterium Infections, Nontuberculous/immunology
- Mycobacterium Infections, Nontuberculous/pathology
- Mycobacterium marinum/isolation & purification
- Mycobacterium marinum/physiology
- Necrosis
- Receptors, Interleukin-4/antagonists & inhibitors
- Receptors, Interleukin-4/genetics
- Receptors, Interleukin-4/metabolism
- STAT6 Transcription Factor/antagonists & inhibitors
- STAT6 Transcription Factor/genetics
- STAT6 Transcription Factor/metabolism
- Signal Transduction
- Zebrafish/growth & development
- Zebrafish/metabolism
- RNA, Guide, CRISPR-Cas Systems
Collapse
Affiliation(s)
- Mark R Cronan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Erika J Hughes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; University Program in Genetics and Genomics, Duke University School of Medicine, Durham, NC 27710, USA
| | - W Jared Brewer
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Gopinath Viswanathan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Emily G Hunt
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Bindu Singh
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Smriti Mehra
- Tulane National Primate Research Center, Covington, LA 70433, USA
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW, Australia; The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW, Australia
| | - Simon G Gregory
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27710, USA
| | - Deepak Kaushal
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
189
|
Wang M, Du L, Lee AC, Li Y, Qin H, He J. Different lineage contexts direct common pro-neural factors to specify distinct retinal cell subtypes. J Cell Biol 2021; 219:151968. [PMID: 32699896 PMCID: PMC7480095 DOI: 10.1083/jcb.202003026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/13/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023] Open
Abstract
How astounding neuronal diversity arises from variable cell lineages in vertebrates remains mostly elusive. By in vivo lineage tracing of ∼1,000 single zebrafish retinal progenitors, we identified a repertoire of subtype-specific stereotyped neurogenic lineages. Remarkably, within these stereotyped lineages, GABAergic amacrine cells were born with photoreceptor cells, whereas glycinergic amacrine cells were born with OFF bipolar cells. More interestingly, post-mitotic differentiation blockage of GABAergic and glycinergic amacrine cells resulted in their respecification into photoreceptor and bipolar cells, respectively, suggesting lineage constraint in cell subtype specification. Using single-cell RNA-seq and ATAC-seq analyses, we further identified lineage-specific progenitors, each defined by specific transcription factors that exhibited characteristic chromatin accessibility dynamics. Finally, single pro-neural factors could specify different neuron types/subtypes in a lineage-dependent manner. Our findings reveal the importance of lineage context in defining neuronal subtypes and provide a demonstration of in vivo lineage-dependent induction of unique retinal neuron subtypes for treatment purposes.
Collapse
Affiliation(s)
- Mei Wang
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Lei Du
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Aih Cheun Lee
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yan Li
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Huiwen Qin
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Jie He
- State Key Laboratory of Neuroscience, Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| |
Collapse
|
190
|
Klatt Shaw D, Mokalled MH. Efficient CRISPR/Cas9 mutagenesis for neurobehavioral screening in adult zebrafish. G3-GENES GENOMES GENETICS 2021; 11:6179145. [PMID: 33742663 PMCID: PMC8496216 DOI: 10.1093/g3journal/jkab089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/07/2021] [Indexed: 12/22/2022]
Abstract
Adult zebrafish are widely used to interrogate mechanisms of disease development and tissue regeneration. Yet, the prospect of large-scale genetics in adult zebrafish has traditionally faced a host of biological and technical challenges, including inaccessibility of adult tissues to high-throughput phenotyping and the spatial and technical demands of adult husbandry. Here, we describe an experimental pipeline that combines high-efficiency CRISPR/Cas9 mutagenesis with functional phenotypic screening to identify genes required for spinal cord repair in adult zebrafish. Using CRISPR/Cas9 dual-guide ribonucleic proteins, we show selective and combinatorial mutagenesis of 17 genes at 28 target sites with efficiencies exceeding 85% in adult F0 “crispants”. We find that capillary electrophoresis is a reliable method to measure indel frequencies. Using a quantifiable behavioral assay, we identify seven single- or duplicate-gene crispants with reduced functional recovery after spinal cord injury. To rule out off-target effects, we generate germline mutations that recapitulate the crispant regeneration phenotypes. This study provides a platform that combines high-efficiency somatic mutagenesis with a functional phenotypic readout to perform medium- to large-scale genetic studies in adult zebrafish.
Collapse
Affiliation(s)
- Dana Klatt Shaw
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA.,Center of Regenerative Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Mayssa H Mokalled
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA.,Center of Regenerative Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
191
|
Sharma P, Sharma BS, Verma RJ. CRISPR-based genome editing of zebrafish. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 180:69-84. [PMID: 33934838 DOI: 10.1016/bs.pmbts.2021.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
CRISPR/Cas9, once discovered as an adaptive immune system in bacteria, has emerged as a disruptive technology in the field of genetic engineering. Technological advancements in the recent past has enhanced the applicability of CRISPR/Cas9 tool for gene editing, gene therapies, developmental studies and mutational analysis in various model organisms. Zebrafish, one of the excellent animal models, is preferred for conducting CRISPR/Cas9 studies to assess the functional implication of specific genes of interest. CRISPR/Cas9 mediated gene editing techniques, such as, knock-out and knock-in approaches, provide evidences to identify the role of different genes through loss-of-function studies. Also, CRISPR/Cas9 has been proved to be an efficient tool for designing disease models for gene expression studies based on phenotypic screening. The present chapter provides an overview of CRISPR/Cas9 mechanism, different strategies for DNA modifications and gene function analysis, highlighting the translational applications for future prospects, such as screening of drug toxicity and efficacy.
Collapse
Affiliation(s)
- Preeti Sharma
- Department of Zoology, Biomedical Technology & Human Genetics, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, India; PanGenomics International Pvt Ltd, Sterling Accuris Diagnostics, Ellis Bridge, Ahmedabad, Gujarat, India.
| | - B Sharan Sharma
- Rivaara Labs Pvt Ltd, KD Hospital, Vaishnodevi Circle, Ahmedabad, Gujarat, India
| | - Ramtej J Verma
- Department of Zoology, Biomedical Technology & Human Genetics, University School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| |
Collapse
|
192
|
Chen W, Zhao J, Mu D, Wang Z, Liu Q, Zhang Y, Yang D. Pyroptosis Mediates Neutrophil Extracellular Trap Formation during Bacterial Infection in Zebrafish. THE JOURNAL OF IMMUNOLOGY 2021; 206:1913-1922. [PMID: 33712519 DOI: 10.4049/jimmunol.2001335] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/05/2021] [Indexed: 12/15/2022]
Abstract
The formation of neutrophil extracellular trap (NET) is a critical host defense when neutrophils migrate to infection sites. Pyroptosis is a newly identified programmed cell death, which is tightly regulated by inflammasome activation. However, the mechanism of pyroptotic signaling participating in NET production remains to be elucidated. In this study, the zebrafish larvae otic vesicle microinjection model was used to infect larvae with hemolysin-overexpressing Edwardsiella piscicida (EthA+), and a rapid migration of neutrophils to infection sites was observed. Intriguingly, EthA+ infection effectively induced significant neutrophil membrane rupture in vivo, which was dependent on caspase-B (caspy2) and gasdermin Eb (GSDMEb) but not caspase-A or gasdermin Ea. Specifically, the EthA+ E. piscicida infection induced pyroptosis along with NETosis in vitro, and depletion of either caspy2 or GSDMEb impaired NET formation in vivo. Consequently, inhibition of the caspy2-GSDMEb axis-gated NETosis impaired bacterial clearance in vivo. Altogether, these data provide evidence that teleost fish innate immune cells, including neutrophils, express features of pyroptosis that are critical for NETosis in teleost innate immunity.
Collapse
Affiliation(s)
- Weijie Chen
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Jingjing Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Di Mu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Zhuang Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, China; and
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.,Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| | - Dahai Yang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; .,Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| |
Collapse
|
193
|
Duan A, Wang H, Zhu Y, Wang Q, Zhang J, Hou Q, Xing Y, Shi J, Hou J, Qin Z, Chen Z, Liu Z, Yang J. Chromatin architecture reveals cell type-specific target genes for kidney disease risk variants. BMC Biol 2021; 19:38. [PMID: 33627123 PMCID: PMC7905576 DOI: 10.1186/s12915-021-00977-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/08/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Cell type-specific transcriptional programming results from the combinatorial interplay between the repertoire of active regulatory elements. Disease-associated variants disrupt such programming, leading to altered expression of downstream regulated genes and the onset of pathological states. However, due to the non-linear regulatory properties of non-coding elements such as enhancers, which can activate transcription at long distances and in a non-directional way, the identification of causal variants and their target genes remains challenging. Here, we provide a multi-omics analysis to identify regulatory elements associated with functional kidney disease variants, and downstream regulated genes. RESULTS In order to understand the genetic risk of kidney diseases, we generated a comprehensive dataset of the chromatin landscape of human kidney tubule cells, including transcription-centered 3D chromatin organization, histone modifications distribution and transcriptome with HiChIP, ChIP-seq and RNA-seq. We identified genome-wide functional elements and thousands of interactions between the distal elements and target genes. The results revealed that risk variants for renal tumor and chronic kidney disease were enriched in kidney tubule cells. We further pinpointed the target genes for the variants and validated two target genes by CRISPR/Cas9 genome editing techniques in zebrafish, demonstrating that SLC34A1 and MTX1 were indispensable genes to maintain kidney function. CONCLUSIONS Our results provide a valuable multi-omics resource on the chromatin landscape of human kidney tubule cells and establish a bioinformatic pipeline in dissecting functions of kidney disease-associated variants based on cell type-specific epigenome.
Collapse
Affiliation(s)
- Aiping Duan
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Hong Wang
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Yan Zhu
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Qi Wang
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Jing Zhang
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Qing Hou
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Yuexian Xing
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Jinsong Shi
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Jinhua Hou
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Zhaohui Qin
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, 1518 Clifton Road N.E, Atlanta, GA, 30322, USA
| | - Zhaohong Chen
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Zhihong Liu
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China
| | - Jingping Yang
- National Clinical Research Center for Kidney Disease, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China.
- Medical School of Nanjing University, Nanjing, 210093, Jiangsu, China.
| |
Collapse
|
194
|
Hans S, Zöller D, Hammer J, Stucke J, Spieß S, Kesavan G, Kroehne V, Eguiguren JS, Ezhkova D, Petzold A, Dahl A, Brand M. Cre-Controlled CRISPR mutagenesis provides fast and easy conditional gene inactivation in zebrafish. Nat Commun 2021; 12:1125. [PMID: 33602923 PMCID: PMC7893016 DOI: 10.1038/s41467-021-21427-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Conditional gene inactivation is a powerful tool to determine gene function when constitutive mutations result in detrimental effects. The most commonly used technique to achieve conditional gene inactivation employs the Cre/loxP system and its ability to delete DNA sequences flanked by two loxP sites. However, targeting a gene with two loxP sites is time and labor consuming. Here, we show Cre-Controlled CRISPR (3C) mutagenesis to circumvent these issues. 3C relies on gRNA and Cre-dependent Cas9-GFP expression from the same transgene. Exogenous or transgenic supply of Cre results in Cas9-GFP expression and subsequent mutagenesis of the gene of interest. The recombined cells become fluorescently visible enabling their isolation and subjection to various omics techniques. Hence, 3C mutagenesis provides a valuable alternative to the production of loxP-flanked alleles. It might even enable the conditional inactivation of multiple genes simultaneously and should be applicable to other model organisms amenable to single integration transgenesis.
Collapse
Affiliation(s)
- Stefan Hans
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.
| | - Daniela Zöller
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Juliane Hammer
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Johanna Stucke
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Sandra Spieß
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Gokul Kesavan
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Volker Kroehne
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Juan Sebastian Eguiguren
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Diana Ezhkova
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany
| | - Andreas Petzold
- Center for Molecular and Cellular Bioengineering (CMCB), DRESDEN-Concept Genome Center, Technische Universität Dresden, Dresden, Germany
| | - Andreas Dahl
- Center for Molecular and Cellular Bioengineering (CMCB), DRESDEN-Concept Genome Center, Technische Universität Dresden, Dresden, Germany
| | - Michael Brand
- Center for Molecular and Cellular Bioengineering (CMCB), Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
195
|
Tobias JH, Duncan EL, Kague E, Hammond CL, Gregson CL, Bassett D, Williams GR, Min JL, Gaunt TR, Karasik D, Ohlsson C, Rivadeneira F, Edwards JR, Hannan FM, Kemp JP, Gilbert SJ, Alonso N, Hassan N, Compston JE, Ralston SH. Opportunities and Challenges in Functional Genomics Research in Osteoporosis: Report From a Workshop Held by the Causes Working Group of the Osteoporosis and Bone Research Academy of the Royal Osteoporosis Society on October 5th 2020. Front Endocrinol (Lausanne) 2021; 11:630875. [PMID: 33658983 PMCID: PMC7917291 DOI: 10.3389/fendo.2020.630875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
The discovery that sclerostin is the defective protein underlying the rare heritable bone mass disorder, sclerosteosis, ultimately led to development of anti-sclerostin antibodies as a new treatment for osteoporosis. In the era of large scale GWAS, many additional genetic signals associated with bone mass and related traits have since been reported. However, how best to interrogate these signals in order to identify the underlying gene responsible for these genetic associations, a prerequisite for identifying drug targets for further treatments, remains a challenge. The resources available for supporting functional genomics research continues to expand, exemplified by "multi-omics" database resources, with improved availability of datasets derived from bone tissues. These databases provide information about potential molecular mediators such as mRNA expression, protein expression, and DNA methylation levels, which can be interrogated to map genetic signals to specific genes based on identification of causal pathways between the genetic signal and the phenotype being studied. Functional evaluation of potential causative genes has been facilitated by characterization of the "osteocyte signature", by broad phenotyping of knockout mice with deletions of over 7,000 genes, in which more detailed skeletal phenotyping is currently being undertaken, and by development of zebrafish as a highly efficient additional in vivo model for functional studies of the skeleton. Looking to the future, this expanding repertoire of tools offers the hope of accurately defining the major genetic signals which contribute to osteoporosis. This may in turn lead to the identification of additional therapeutic targets, and ultimately new treatments for osteoporosis.
Collapse
Affiliation(s)
- Jonathan H. Tobias
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Emma L. Duncan
- Department of Twin Research & Genetic Epidemiology, School of Life Course Sciences, King’s College London, London, United Kingdom
| | - Erika Kague
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Chrissy L. Hammond
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, United Kingdom
| | - Celia L. Gregson
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion & Reproduction, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Graham R. Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion & Reproduction, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | - Josine L. Min
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Tom R. Gaunt
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Claes Ohlsson
- Center for Bone and Arthritis Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - James R. Edwards
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Fadil M. Hannan
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - John P. Kemp
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- University of Queensland Diamantina Institute, University of Queensland, Woolloongabba, Queensland, QLD, Australia
| | - Sophie J. Gilbert
- Biomechanics and Bioengineering Centre Versus Arthritis, Cardiff School of Biosciences, Cardiff, United Kingdom
| | - Nerea Alonso
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Neelam Hassan
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Juliet E. Compston
- Department of Medicine, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Stuart H. Ralston
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
196
|
Kemmler CL, Riemslagh FW, Moran HR, Mosimann C. From Stripes to a Beating Heart: Early Cardiac Development in Zebrafish. J Cardiovasc Dev Dis 2021; 8:17. [PMID: 33578943 PMCID: PMC7916704 DOI: 10.3390/jcdd8020017] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 12/18/2022] Open
Abstract
The heart is the first functional organ to form during vertebrate development. Congenital heart defects are the most common type of human birth defect, many originating as anomalies in early heart development. The zebrafish model provides an accessible vertebrate system to study early heart morphogenesis and to gain new insights into the mechanisms of congenital disease. Although composed of only two chambers compared with the four-chambered mammalian heart, the zebrafish heart integrates the core processes and cellular lineages central to cardiac development across vertebrates. The rapid, translucent development of zebrafish is amenable to in vivo imaging and genetic lineage tracing techniques, providing versatile tools to study heart field migration and myocardial progenitor addition and differentiation. Combining transgenic reporters with rapid genome engineering via CRISPR-Cas9 allows for functional testing of candidate genes associated with congenital heart defects and the discovery of molecular causes leading to observed phenotypes. Here, we summarize key insights gained through zebrafish studies into the early patterning of uncommitted lateral plate mesoderm into cardiac progenitors and their regulation. We review the central genetic mechanisms, available tools, and approaches for modeling congenital heart anomalies in the zebrafish as a representative vertebrate model.
Collapse
Affiliation(s)
| | | | | | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine and Children’s Hospital Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA; (C.L.K.); (F.W.R.); (H.R.M.)
| |
Collapse
|
197
|
Kroll F, Powell GT, Ghosh M, Gestri G, Antinucci P, Hearn TJ, Tunbak H, Lim S, Dennis HW, Fernandez JM, Whitmore D, Dreosti E, Wilson SW, Hoffman EJ, Rihel J. A simple and effective F0 knockout method for rapid screening of behaviour and other complex phenotypes. eLife 2021; 10:e59683. [PMID: 33416493 PMCID: PMC7793621 DOI: 10.7554/elife.59683] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
Hundreds of human genes are associated with neurological diseases, but translation into tractable biological mechanisms is lagging. Larval zebrafish are an attractive model to investigate genetic contributions to neurological diseases. However, current CRISPR-Cas9 methods are difficult to apply to large genetic screens studying behavioural phenotypes. To facilitate rapid genetic screening, we developed a simple sequencing-free tool to validate gRNAs and a highly effective CRISPR-Cas9 method capable of converting >90% of injected embryos directly into F0 biallelic knockouts. We demonstrate that F0 knockouts reliably recapitulate complex mutant phenotypes, such as altered molecular rhythms of the circadian clock, escape responses to irritants, and multi-parameter day-night locomotor behaviours. The technique is sufficiently robust to knockout multiple genes in the same animal, for example to create the transparent triple knockout crystal fish for imaging. Our F0 knockout method cuts the experimental time from gene to behavioural phenotype in zebrafish from months to one week.
Collapse
Affiliation(s)
- François Kroll
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Gareth T Powell
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Marcus Ghosh
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Gaia Gestri
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Paride Antinucci
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| | - Timothy J Hearn
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Hande Tunbak
- Wolfson Institute for Biomedical Research, University College LondonLondonUnited Kingdom
| | - Sumi Lim
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Harvey W Dennis
- School of Biological Sciences, Faculty of Science, University of BristolBristolUnited Kingdom
| | | | - David Whitmore
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
- Department of Molecular and Cell Biology, James Cook UniversityTownsvilleAustralia
| | - Elena Dreosti
- Wolfson Institute for Biomedical Research, University College LondonLondonUnited Kingdom
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Ellen J Hoffman
- Child Study Center, Yale School of MedicineNew HavenUnited States
- Department of Neuroscience, Yale School of MedicineNew HavenUnited States
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| |
Collapse
|
198
|
Zhao D, Jones JL, Gasperini RJ, Charlesworth JC, Liu GS, Burdon KP. Rapid and efficient cataract gene evaluation in F0 zebrafish using CRISPR-Cas9 ribonucleoprotein complexes. Methods 2021; 194:37-47. [PMID: 33418061 DOI: 10.1016/j.ymeth.2020.12.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/22/2020] [Accepted: 12/29/2020] [Indexed: 02/08/2023] Open
Abstract
Cataract is the leading cause of blindness worldwide. Congenital or paediatric cataract can result in permanent visual impairment or blindness even with best attempts at treatment. A significant proportion of paediatric cataract has a genetic cause. Therefore, identifying the genes that lead to cataract formation is essential for understanding the pathological process of inherited paediatric cataract as well as to the development of new therapies. Despite clear progress in genomics technologies, verification of the biological effects of newly identified candidate genes and variants is still challenging. Here, we provide a step-by-step pipeline to evaluate cataract candidate genes in F0 zebrafish using CRISPR-Cas9 ribonucleoprotein complexes (RNP). Detailed descriptions of CRISPR-Cas9 RNP design and formulation, microinjection, optimization of CRISPR-Cas9 RNP reagent dose and delivery route, editing efficacy analysis as well as cataract formation evaluation are included. Following this protocol, any cataract candidates can be readily and efficiently evaluated within 2 weeks using basic laboratory supplies.
Collapse
Affiliation(s)
- Duran Zhao
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Johanna L Jones
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | | | - Jac C Charlesworth
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Guei-Sheung Liu
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, Victoria, Australia
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Department of Ophthalmology, Flinders University, Bedford Park, South Australia, Australia.
| |
Collapse
|
199
|
Sieliwonczyk E, Matchkov VV, Vandendriessche B, Alaerts M, Bakkers J, Loeys B, Schepers D. Inherited Ventricular Arrhythmia in Zebrafish: Genetic Models and Phenotyping Tools. Rev Physiol Biochem Pharmacol 2021; 184:33-68. [PMID: 34533615 DOI: 10.1007/112_2021_65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
In the last years, the field of inheritable ventricular arrhythmia disease modelling has changed significantly with a push towards the use of novel cellular cardiomyocyte based models. However, there is a growing need for new in vivo models to study the disease pathology at the tissue and organ level. Zebrafish provide an excellent opportunity for in vivo modelling of inheritable ventricular arrhythmia syndromes due to the remarkable similarity between their cardiac electrophysiology and that of humans. Additionally, many state-of-the-art methods in gene editing and electrophysiological phenotyping are available for zebrafish research. In this review, we give a comprehensive overview of the published zebrafish genetic models for primary electrical disorders and arrhythmogenic cardiomyopathy. We summarise and discuss the strengths and weaknesses of the different technical approaches for the generation of genetically modified zebrafish disease models, as well as the electrophysiological approaches in zebrafish phenotyping. By providing this detailed overview, we aim to draw attention to the potential of the zebrafish model for studying arrhythmia syndromes at the organ level and as a platform for personalised medicine and drug testing.
Collapse
Affiliation(s)
- Ewa Sieliwonczyk
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.
| | - Vladimir V Matchkov
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Bert Vandendriessche
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Maaike Alaerts
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Jeroen Bakkers
- Hubrecht Institute for Developmental and Stem Cell Biology, Utrecht, The Netherlands
| | - Bart Loeys
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Dorien Schepers
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium.,Laboratory for Molecular, Cellular and Network Excitability, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
200
|
Abstract
5-methylcytosine (5mC) is a gene-regulatory mark associated with transcriptional repression. 5mC can be erased through the catalytic action of Ten-eleven translocation (TET) methylcytosine dioxygenases (TET1, TET2, TET3), which oxidize 5mC resulting in its removal from the genome. In vertebrates, TET enzymes facilitate DNA demethylation of regulatory regions linked to genes involved in developmental processes. Consequently, TET ablation leads to severe morphological defects and developmental arrest. Here we describe a system that can facilitate the study of relationships between TET enzymes, 5mC, and embryo development. We provide detailed descriptions for the generation of F0 zebrafish tet1/2/3 knockouts using CRISPR/Cas9 technology and elaborate on the strategies to assess the impact of TET loss by reduced representation bisulfite sequencing (RRBS).
Collapse
|