151
|
Vaz R, Hofmeister W, Lindstrand A. Zebrafish Models of Neurodevelopmental Disorders: Limitations and Benefits of Current Tools and Techniques. Int J Mol Sci 2019; 20:ijms20061296. [PMID: 30875831 PMCID: PMC6471844 DOI: 10.3390/ijms20061296] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/26/2019] [Accepted: 03/11/2019] [Indexed: 12/13/2022] Open
Abstract
For the past few years there has been an exponential increase in the use of animal models to confirm the pathogenicity of candidate disease-causing genetic variants found in patients. One such animal model is the zebrafish. Despite being a non-mammalian animal, the zebrafish model has proven its potential in recapitulating the phenotypes of many different human genetic disorders. This review will focus on recent advances in the modeling of neurodevelopmental disorders in zebrafish, covering aspects from early brain development to techniques used for modulating gene expression, as well as how to best characterize the resulting phenotypes. We also review other existing models of neurodevelopmental disorders, and the current efforts in developing and testing compounds with potential therapeutic value.
Collapse
Affiliation(s)
- Raquel Vaz
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden.
| | - Wolfgang Hofmeister
- Laboratory of Molecular and Cellular Cardiology, Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, 5000 Odense, Denmark and the Novo Nordisk Foundation for Stem cell Biology (Danstem), University of Copenhagen, 2200 Copenhagen, Denmark.
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Center for Molecular Medicine and Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, 171 76 Stockholm, Sweden.
| |
Collapse
|
152
|
Tabor KM, Marquart GD, Hurt C, Smith TS, Geoca AK, Bhandiwad AA, Subedi A, Sinclair JL, Rose HM, Polys NF, Burgess HA. Brain-wide cellular resolution imaging of Cre transgenic zebrafish lines for functional circuit-mapping. eLife 2019; 8:42687. [PMID: 30735129 PMCID: PMC6392497 DOI: 10.7554/elife.42687] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/07/2019] [Indexed: 12/14/2022] Open
Abstract
Decoding the functional connectivity of the nervous system is facilitated by transgenic methods that express a genetically encoded reporter or effector in specific neurons; however, most transgenic lines show broad spatiotemporal and cell-type expression. Increased specificity can be achieved using intersectional genetic methods which restrict reporter expression to cells that co-express multiple drivers, such as Gal4 and Cre. To facilitate intersectional targeting in zebrafish, we have generated more than 50 new Cre lines, and co-registered brain expression images with the Zebrafish Brain Browser, a cellular resolution atlas of 264 transgenic lines. Lines labeling neurons of interest can be identified using a web-browser to perform a 3D spatial search (zbbrowser.com). This resource facilitates the design of intersectional genetic experiments and will advance a wide range of precision circuit-mapping studies.
Collapse
Affiliation(s)
- Kathryn M Tabor
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States
| | - Gregory D Marquart
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States.,Neuroscience and Cognitive Science Program, University of Maryland, College Park, United States
| | - Christopher Hurt
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States.,Advanced Research Computing, Department of Computer Science, Virginia Polytechnic Institute and State University, Blacksburg, United States
| | - Trevor S Smith
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States
| | - Alexandra K Geoca
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States
| | - Ashwin A Bhandiwad
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States
| | - Abhignya Subedi
- Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, Bethesda, United States
| | - Jennifer L Sinclair
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States
| | - Hannah M Rose
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States
| | - Nicholas F Polys
- Advanced Research Computing, Department of Computer Science, Virginia Polytechnic Institute and State University, Blacksburg, United States
| | - Harold A Burgess
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, United States
| |
Collapse
|
153
|
Han Y, Chen A, Umansky KB, Oonk KA, Choi WY, Dickson AL, Ou J, Cigliola V, Yifa O, Cao J, Tornini VA, Cox BD, Tzahor E, Poss KD. Vitamin D Stimulates Cardiomyocyte Proliferation and Controls Organ Size and Regeneration in Zebrafish. Dev Cell 2019; 48:853-863.e5. [PMID: 30713073 DOI: 10.1016/j.devcel.2019.01.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/15/2018] [Accepted: 12/28/2018] [Indexed: 01/07/2023]
Abstract
Attaining proper organ size during development and regeneration hinges on the activity of mitogenic factors. Here, we performed a large-scale chemical screen in embryonic zebrafish to identify cardiomyocyte mitogens. Although commonly considered anti-proliferative, vitamin D analogs like alfacalcidol had rapid, potent mitogenic effects on embryonic and adult cardiomyocytes in vivo. Moreover, pharmacologic or genetic manipulation of vitamin D signaling controlled proliferation in multiple adult cell types and dictated growth rates in embryonic and juvenile zebrafish. Tissue-specific modulation of vitamin D receptor (VDR) signaling had organ-restricted effects, with cardiac VDR activation causing cardiomegaly. Alfacalcidol enhanced the regenerative response of injured zebrafish hearts, whereas VDR blockade inhibited regeneration. Alfacalcidol activated cardiac expression of genes associated with ErbB2 signaling, while ErbB2 inhibition blunted its effects on cell proliferation. Our findings identify vitamin D as mitogenic for cardiomyocytes and other cell types in zebrafish and indicate a mechanism to regulate organ size and regeneration.
Collapse
Affiliation(s)
- Yanchao Han
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Anzhi Chen
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Kfir-Baruch Umansky
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Kelsey A Oonk
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Wen-Yee Choi
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Amy L Dickson
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Jianhong Ou
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Valentina Cigliola
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Oren Yifa
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Jingli Cao
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Valerie A Tornini
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Ben D Cox
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA
| | - Eldad Tzahor
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Kenneth D Poss
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Regeneration Next, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
154
|
Ando K, Wang W, Peng D, Chiba A, Lagendijk AK, Barske L, Crump JG, Stainier DYR, Lendahl U, Koltowska K, Hogan BM, Fukuhara S, Mochizuki N, Betsholtz C. Peri-arterial specification of vascular mural cells from naïve mesenchyme requires Notch signaling. Development 2019; 146:dev.165589. [PMID: 30642834 DOI: 10.1242/dev.165589] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 12/21/2018] [Indexed: 12/30/2022]
Abstract
Mural cells (MCs) are essential for blood vessel stability and function; however, the mechanisms that regulate MC development remain incompletely understood, in particular those involved in MC specification. Here, we investigated the first steps of MC formation in zebrafish using transgenic reporters. Using pdgfrb and abcc9 reporters, we show that the onset of expression of abcc9, a pericyte marker in adult mice and zebrafish, occurs almost coincidentally with an increment in pdgfrb expression in peri-arterial mesenchymal cells, suggesting that these transcriptional changes mark the specification of MC lineage cells from naïve pdgfrb low mesenchymal cells. The emergence of peri-arterial pdgfrb high MCs required Notch signaling. We found that pdgfrb-positive cells express notch2 in addition to notch3, and although depletion of notch2 or notch3 failed to block MC emergence, embryos depleted of both notch2 and notch3 lost mesoderm- as well as neural crest-derived pdgfrb high MCs. Using reporters that read out Notch signaling and Notch2 receptor cleavage, we show that Notch activation in the mesenchyme precedes specification into pdgfrb high MCs. Taken together, these results show that Notch signaling is necessary for peri-arterial MC specification.
Collapse
Affiliation(s)
- Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden .,Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565 8565, Japan
| | - Weili Wang
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Di Peng
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Ayano Chiba
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565 8565, Japan
| | - Anne K Lagendijk
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Lindsey Barske
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Biomedicum, Solnavägen 9, SE-171 77 Stockholm, Sweden.,Department of Medicine, Huddinge, Karolinska Institutet, Integrated Cardio Metabolic Centre (ICMC), Blickagången 6, SE-141 57 Huddinge, Sweden
| | - Katarzyna Koltowska
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden.,Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, Queensland 4072, Australia
| | - Shigetomo Fukuhara
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565 8565, Japan.,Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School Musashi Kosugi Hospital, Kawasaki, Kanagawa 211 8533, Japan
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565 8565, Japan.,AMED-CREST, Department of Cell Biology, National Cerebral and Cardiovascular Center, 5-7-1, Suita, Osaka 565 8565, Japan
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden.,Department of Medicine, Huddinge, Karolinska Institutet, Integrated Cardio Metabolic Centre (ICMC), Blickagången 6, SE-141 57 Huddinge, Sweden
| |
Collapse
|
155
|
Knogler LD, Kist AM, Portugues R. Motor context dominates output from purkinje cell functional regions during reflexive visuomotor behaviours. eLife 2019; 8:e42138. [PMID: 30681408 PMCID: PMC6374073 DOI: 10.7554/elife.42138] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/26/2018] [Indexed: 12/22/2022] Open
Abstract
The cerebellum integrates sensory stimuli and motor actions to enable smooth coordination and motor learning. Here we harness the innate behavioral repertoire of the larval zebrafish to characterize the spatiotemporal dynamics of feature coding across the entire Purkinje cell population during visual stimuli and the reflexive behaviors that they elicit. Population imaging reveals three spatially-clustered regions of Purkinje cell activity along the rostrocaudal axis. Complementary single-cell electrophysiological recordings assign these Purkinje cells to one of three functional phenotypes that encode a specific visual, and not motor, signal via complex spikes. In contrast, simple spike output of most Purkinje cells is strongly driven by motor-related tail and eye signals. Interactions between complex and simple spikes show heterogeneous modulation patterns across different Purkinje cells, which become temporally restricted during swimming episodes. Our findings reveal how sensorimotor information is encoded by individual Purkinje cells and organized into behavioral modules across the entire cerebellum.
Collapse
Affiliation(s)
- Laura D Knogler
- Max Planck Institute of Neurobiology, Sensorimotor Control Research GroupMartinsriedGermany
| | - Andreas M Kist
- Max Planck Institute of Neurobiology, Sensorimotor Control Research GroupMartinsriedGermany
| | - Ruben Portugues
- Max Planck Institute of Neurobiology, Sensorimotor Control Research GroupMartinsriedGermany
| |
Collapse
|
156
|
Gunawan F, Gentile A, Fukuda R, Tsedeke AT, Jiménez-Amilburu V, Ramadass R, Iida A, Sehara-Fujisawa A, Stainier DYR. Focal adhesions are essential to drive zebrafish heart valve morphogenesis. J Cell Biol 2019; 218:1039-1054. [PMID: 30635353 PMCID: PMC6400548 DOI: 10.1083/jcb.201807175] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/07/2018] [Accepted: 12/13/2018] [Indexed: 12/17/2022] Open
Abstract
Gunawan et al. analyze at single-cell resolution collective endocardial cell migration into the extracellular matrix and the cellular rearrangements forming leaflets during zebrafish heart valve formation. They show that focal adhesion activity driven by Integrin α5β1 and Talin1 are essential to drive cardiac valve morphogenesis in zebrafish. Elucidating the morphogenetic events that shape vertebrate heart valves, complex structures that prevent retrograde blood flow, is critical to understanding valvular development and aberrations. Here, we used the zebrafish atrioventricular (AV) valve to investigate these events in real time and at single-cell resolution. We report the initial events of collective migration of AV endocardial cells (ECs) into the extracellular matrix (ECM), and their subsequent rearrangements to form the leaflets. We functionally characterize integrin-based focal adhesions (FAs), critical mediators of cell–ECM interactions, during valve morphogenesis. Using transgenes to block FA signaling specifically in AV ECs as well as loss-of-function approaches, we show that FA signaling mediated by Integrin α5β1 and Talin1 promotes AV EC migration and overall shaping of the valve leaflets. Altogether, our investigation reveals the critical processes driving cardiac valve morphogenesis in vivo and establishes the zebrafish AV valve as a vertebrate model to study FA-regulated tissue morphogenesis.
Collapse
Affiliation(s)
- Felix Gunawan
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Alessandra Gentile
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ryuichi Fukuda
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ayele Taddese Tsedeke
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Vanesa Jiménez-Amilburu
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Radhan Ramadass
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Atsuo Iida
- Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | | | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
157
|
Zhang Y, Ouyang J, Qie J, Zhang G, Liu L, Yang P. Optimization of the Gal4/UAS transgenic tools in zebrafish. Appl Microbiol Biotechnol 2019; 103:1789-1799. [PMID: 30613898 DOI: 10.1007/s00253-018-09591-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/14/2018] [Accepted: 12/20/2018] [Indexed: 10/27/2022]
Abstract
The Gal4/UAS system provides a powerful tool to analyze the function of genes. The system has been employed extensively in zebrafish; however, cytotoxicity of Gal4 and methylation of UAS can hinder future applications of Gal4/UAS in zebrafish. In this study, we provide quantitative data on the cytotoxicity of Gal4-FF and KalTA4 in zebrafish embryos. A better balance between induction efficiency and toxicity was shown when the injection dosage was 20 pg for Gal4-FF and 30 pg for KalTA4. We tested the DNA methylation of UAS in different copies (3×, 5×, 7×, 9×, 11×, and 14×), and the results showed, for the first time, that the degree of UAS methylation increases with the increase in the copy number of UAS. We detected insertions of the Tol2-mediated transgene in the Gal4 line and found as many as three sites of insertion, on average; only about 20% of individuals contained single-site insertion in F1 generation. We suggested that the screening of Gal4 lines with single-site insertion is essential when Tol2-mediated Gal4 transgenic lines are created. Moreover, we designed a novel 5 × non-repetitive UAS (5 × nrUAS) to reduce the appeal of multicopy UAS as a target for methylation. Excitingly, the 5 × nrUAS is less prone to methylation compared to 5 × UAS. We hope the results will facilitate the future application of the Gal4/UAS system in zebrafish research.
Collapse
Affiliation(s)
- Yunsheng Zhang
- Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Key Laboratory of Health Aquaculture and Product Processing in Dongting Lake Area of Hunan Province, Zoology Key Laboratory of Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, Hunan, People's Republic of China.
| | - Jiawei Ouyang
- Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Key Laboratory of Health Aquaculture and Product Processing in Dongting Lake Area of Hunan Province, Zoology Key Laboratory of Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, Hunan, People's Republic of China
| | - Jingrong Qie
- Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Key Laboratory of Health Aquaculture and Product Processing in Dongting Lake Area of Hunan Province, Zoology Key Laboratory of Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, Hunan, People's Republic of China
| | - Gongyuan Zhang
- Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Key Laboratory of Health Aquaculture and Product Processing in Dongting Lake Area of Hunan Province, Zoology Key Laboratory of Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, Hunan, People's Republic of China
| | - Liangguo Liu
- Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Key Laboratory of Health Aquaculture and Product Processing in Dongting Lake Area of Hunan Province, Zoology Key Laboratory of Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, Hunan, People's Republic of China
| | - Pinhong Yang
- Collaborative Innovation Center for Efficient and Health Production of Fisheries in Hunan Province, Key Laboratory of Health Aquaculture and Product Processing in Dongting Lake Area of Hunan Province, Zoology Key Laboratory of Hunan Higher Education, Hunan University of Arts and Science, Changde, 415000, Hunan, People's Republic of China
| |
Collapse
|
158
|
Enhancer Trapping and Annotation in Zebrafish Mediated with Sleeping Beauty, piggyBac and Tol2 Transposons. Genes (Basel) 2018; 9:genes9120630. [PMID: 30551672 PMCID: PMC6316676 DOI: 10.3390/genes9120630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/07/2018] [Accepted: 12/10/2018] [Indexed: 12/18/2022] Open
Abstract
Although transposon-mediated enhancer trapping (ET) is successfully applied in diverse models, the efficiency of various transposon systems varies significantly, and little information is available regarding efficiency of enhancer trapping by various transposons in zebrafish. Most potential enhancers (Ens) still lack evidence of actual En activity. Here, we compared the differences in ET efficiency between sleeping beauty (SB), piggyBac (PB) and Tol2 transposons. Tol2 represented the highest germline transfer efficiencies at 55.56% (NF0 = 165), followed by SB (38.36%, NF0 = 151) and PB (32.65%, NF0 = 149). ET lines generated by the Tol2 transposon tended to produce offspring with a single expression pattern per line, while PB and SB tended to generate embryos with multiple expression patterns. In our tests, 10 putative Ens (En1–10) were identified by splinkerette PCR and comparative genomic analysis. Combining the GFP expression profiles and mRNA expression patterns revealed that En1 and En2 may be involved in regulation of the expression of dlx1a and dlx2a, while En6 may be involved in regulation of the expression of line TK4 transgene and rps26, and En7 may be involved in the regulation of the expression of wnt1 and wnt10b. Most identified Ens were found to be transcribed in zebrafish embryos, and their regulatory function may involve eRNAs.
Collapse
|
159
|
Audira G, Sarasamma S, Chen JR, Juniardi S, Sampurna BP, Liang ST, Lai YH, Lin GM, Hsieh MC, Hsiao CD. Zebrafish Mutants Carrying Leptin a (lepa) Gene Deficiency Display Obesity, Anxiety, Less Aggression and Fear, and Circadian Rhythm and Color Preference Dysregulation. Int J Mol Sci 2018; 19:ijms19124038. [PMID: 30551684 PMCID: PMC6320766 DOI: 10.3390/ijms19124038] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/05/2018] [Accepted: 12/11/2018] [Indexed: 01/14/2023] Open
Abstract
Leptin, a hormone secreted by peripheral adipose tissues, regulates the appetite in animals. Recently, evidence has shown that leptin also plays roles in behavioral response in addition to controlling appetite. In this study, we examined the potential function of leptin on non-appetite behaviors in zebrafish model. By using genome editing tool of Transcription activator-like effector nuclease (TALEN), we successfully knocked out leptin a (lepa) gene by deleting 4 bp within coding region to create a premature-translation stop. Morphological and appetite analysis showed the lepa KO fish display a phenotype with obese, good appetite and elevation of Agouti-related peptide (AgRP) and Ghrelin hormones, consistent with the canonical function of leptin in controlling food intake. By multiple behavior endpoint analyses, including novel tank, mirror biting, predator avoidance, social interaction, shoaling, circadian rhythm, and color preference assay, we found the lepa KO fish display an anxiogenic phenotype showing hyperactivity with rapid swimming, less freezing time, less fear to predator, loose shoaling area forming, and circadian rhythm and color preference dysregulations. Using biochemical assays, melatonin, norepinephrine, acetylcholine and serotonin levels in the brain were found to be significantly reduced in lepa KO fish, while the levels of dopamine, glycine and cortisol in the brain were significantly elevated. In addition, the brain ROS level was elevated, and the anti-oxidative enzyme catalase level was reduced. Taken together, by performing loss-of-function multiple behavior endpoint testing and biochemical analysis, we provide strong evidence for a critical role of lepa gene in modulating anxiety, aggression, fear, and circadian rhythm behaviors in zebrafish for the first time.
Collapse
Affiliation(s)
- Gilbert Audira
- Department of Chemistry, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| | - Sreeja Sarasamma
- Department of Chemistry, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| | - Jung-Ren Chen
- Department of Biological Science & Technology College of Medicine, I-Shou University, Kaohsiung, 82445, Taiwan.
| | - Stevhen Juniardi
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| | | | - Sung-Tzu Liang
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| | - Yu-Heng Lai
- Department of Chemistry, Chinese Culture University, Taipei 11114, Taiwan.
| | - Geng-Ming Lin
- Laboratory of Marine Biology and Ecology, Third Institute of Oceanography, State OceanicAdministration, Xiamen 361005, China.
| | - Ming-Chia Hsieh
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Changhua Christian Hospital, Changhua 50094, Taiwan.
| | - Chung-Der Hsiao
- Department of Chemistry, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Department of Bioscience Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Center of Nanotechnology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
- Center of Biomedical Technology, Chung Yuan Christian University, Chung-Li 32023, Taiwan.
| |
Collapse
|
160
|
Zhong Y, Huang W, Du J, Wang Z, He J, Luo L. Improved Tol2-mediated enhancer trap identifies weakly expressed genes during liver and β cell development and regeneration in zebrafish. J Biol Chem 2018; 294:932-940. [PMID: 30504219 DOI: 10.1074/jbc.ra118.005568] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/26/2018] [Indexed: 01/13/2023] Open
Abstract
The liver and pancreas are two major digestive organs, and among the different cell types in them, hepatocytes and the insulin-producing β cells have roles in both health and diseases. Accordingly, clinicians and researchers are very interested in the mechanisms underlying the development and regeneration of liver and pancreatic β cells. Gene and enhancer traps such as the Tol2 transposon-based system are useful for identifying genes potentially involved in developmental processes in the zebrafish model. Here, we developed a strategy that combines a Tol2-mediated enhancer trap and the Cre/loxP system by using loxP-flanked reporters driven by β cell- or hepatocyte-specific promoters and the upstream activating sequence (UAS)-driving Cre. Two double-transgenic reporter lines, Tg(ins:loxP-CFPNTR-loxP-DsRed; 10×UAS:Cre, cryaa:Venus) and Tg(fabp10:loxP-CFPNTR-loxP-DsRed; 10×UAS:Cre, cryaa:Venus), were established to label pancreatic β cells and hepatocytes, respectively. These two double-transgenic lines were each crossed with the Tol2-enhancer trap founder lines to screen for and identify genes expressed in the β cell and hepatocytes during development. This trap system coupled with application of nitroreductase (NTR)/metronidazole (Mtz)-mediated cell ablation could identify genes expressed during regeneration. Of note, pilot enhancer traps captured transiently and weakly expressed genes such as rab3da and ensab with higher efficiencies than traditional enhancer trap systems. In conclusion, through permanent genetic labeling by Cre/loxP, this improved Tol2-mediated enhancer trap system provides a promising method to identify transiently or weakly expressed, but potentially important, genes during development and regeneration.
Collapse
Affiliation(s)
- Yadong Zhong
- From the Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, and.,Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Wei Huang
- From the Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, and.,Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Jiang Du
- From the Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, and.,Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Zekun Wang
- From the Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, and.,Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Jianbo He
- From the Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, and.,Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| | - Lingfei Luo
- From the Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, and .,Laboratory of Molecular Developmental Biology, School of Life Sciences, Southwest University, Beibei, 400715 Chongqing, China
| |
Collapse
|
161
|
Optogenetic precision toolkit to reveal form, function and connectivity of single neurons. Methods 2018; 150:42-48. [DOI: 10.1016/j.ymeth.2018.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/24/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
|
162
|
Iida A, Wang Z, Hirata H, Sehara-Fujisawa A. Integrin β1 activity is required for cardiovascular formation in zebrafish. Genes Cells 2018; 23:938-951. [PMID: 30151851 DOI: 10.1111/gtc.12641] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022]
Abstract
Integrins are transmembrane molecules that facilitate cell-to-cell and cell-to-extracellular matrix (ECM) interactions. Integrin molecules are heterodimers that consist of α- and β-subunits. The integrin β1 gene is widely expressed in vivo and is the major β molecule in many tissues; however, tissue-specific roles of integrin β1 are still elusive. In this study, we investigated integrin β1 function in endothelial cells of zebrafish. An integrin β1b mutant zebrafish exhibited morphological abnormalities in blood vessel formation, cephalic hemorrhage and a decreased responsiveness to tactile stimulation during development. To determine the role of integrin β1b in vascular formation, we developed a Gal4/UAS-mediated conditional inactivation of integrin β1 by expressing the cytoplasmic region of integrin β1 that acts as a dominant-negative (DN) isoform. Expression of integrin β1 DN in endothelial cells induced blood vessel abnormalities as in integrin β1b mutants. These results show that endothelial cells require integrin activity for the formation and/or maintenance of blood vessels in zebrafish. Furthermore, our time-lapse recording visualized the breakpoint of cephalic vessels and the hemorrhage onset. Taken together, our tissue-specific inactivation of integrin β1 in zebrafish is powerful tools for functional analysis of integrin β1 in developing tissues.
Collapse
Affiliation(s)
- Atsuo Iida
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Zi Wang
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiromi Hirata
- Department of Chemistry and Biological Science, Graduate School of Science and Engineering, Aoyama Gakuin University, Sagamihara, Japan
| | - Atsuko Sehara-Fujisawa
- Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
163
|
Du XF, Xu B, Zhang Y, Chen MJ, Du JL. A transgenic zebrafish model for in vivo long-term imaging of retinotectal synaptogenesis. Sci Rep 2018; 8:14077. [PMID: 30232367 PMCID: PMC6145912 DOI: 10.1038/s41598-018-32409-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/04/2018] [Indexed: 01/07/2023] Open
Abstract
The retinotectal synapse in larval zebrafish, combined with live time-lapse imaging, provides an advantageous model for study of the development and remodelling of central synapses in vivo. In previous studies, these synapses were labelled by transient expression of fluorescence-tagged synaptic proteins, which resulted in the dramatic variation of labelling patterns in each larva. Here, using GAL4-Upstream Activating Sequence (GAL4-UAS) methodology, we generated stable transgenic lines, which express EGFP-tagged synaptophysin (a presynaptic protein) in retinal ganglion cells (RGCs), to reliably label the pre-synaptic site of retinotectal synapses. This tool avoids the variable labelling of RGCs that occurs in transient transgenic larvae. We obtained several stable transgenic lines that differ consistently in the number of labelled RGCs. Using stable lines that consistently had a single labelled RGC, we could trace synaptogenic dynamics on an individual RGC axonal arbor across different developmental stages. In the stable lines that consistently had multiple labelled RGCs, we could simultaneously monitor both pre- and post-synaptic compartments by combining transient labelling of post-synaptic sites on individual tectal neurons. These tools allowed us to investigate molecular events underlying synaptogenesis and found that the microRNA-132 (miR-132) is required for developmental synaptogenesis. Thus, these transgenic zebrafish stable lines provide appropriate tools for studying central synaptogenesis and underlying molecular mechanisms in intact vertebrate brain.
Collapse
Affiliation(s)
- Xu-Fei Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.
| | - Bing Xu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China
| | - Yu Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing, 100049, China
| | - Min-Jia Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, Shanghai, 200031, China
| | - Jiu-Lin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai, 200031, China. .,School of Future Technology, University of Chinese Academy of Sciences, 19A Yu-Quan Road, Beijing, 100049, China. .,School of Life Science and Technology, ShanghaiTech University, 319 Yue-Yang Road, Shanghai, 200031, China.
| |
Collapse
|
164
|
Jardin N, Giudicelli F, Ten Martín D, Vitrac A, De Gois S, Allison R, Houart C, Reid E, Hazan J, Fassier C. BMP- and neuropilin 1-mediated motor axon navigation relies on spastin alternative translation. Development 2018; 145:dev.162701. [PMID: 30082270 PMCID: PMC6141775 DOI: 10.1242/dev.162701] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 07/16/2018] [Indexed: 12/22/2022]
Abstract
Functional analyses of genes responsible for neurodegenerative disorders have unveiled crucial links between neurodegenerative processes and key developmental signalling pathways. Mutations in SPG4-encoding spastin cause hereditary spastic paraplegia (HSP). Spastin is involved in diverse cellular processes that couple microtubule severing to membrane remodelling. Two main spastin isoforms are synthesised from alternative translational start sites (M1 and M87). However, their specific roles in neuronal development and homeostasis remain largely unknown. To selectively unravel their neuronal function, we blocked spastin synthesis from each initiation codon during zebrafish development and performed rescue analyses. The knockdown of each isoform led to different motor neuron and locomotion defects, which were not rescued by the selective expression of the other isoform. Notably, both morphant neuronal phenotypes were observed in a CRISPR/Cas9 spastin mutant. We next showed that M1 spastin, together with HSP proteins atlastin 1 and NIPA1, drives motor axon targeting by repressing BMP signalling, whereas M87 spastin acts downstream of neuropilin 1 to control motor neuron migration. Our data therefore suggest that defective BMP and neuropilin 1 signalling may contribute to the motor phenotype in a vertebrate model of spastin depletion.
Collapse
Affiliation(s)
- Nicolas Jardin
- Sorbonne Universités, UPMC Université Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), 75005 Paris, France
| | - François Giudicelli
- Sorbonne Universités, UPMC Université Paris 06, CNRS, INSERM, Biologie du Développement Paris Seine - Institut de Biologie Paris-Seine (LBD-IBPS), 75005 Paris, France
| | - Daniel Ten Martín
- Sorbonne Universités, UPMC Université Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), 75005 Paris, France
| | - Anaïs Vitrac
- Sorbonne Universités, UPMC Université Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), 75005 Paris, France
| | - Stéphanie De Gois
- Sorbonne Universités, UPMC Université Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), 75005 Paris, France
| | - Rachel Allison
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 OXY, UK
| | - Corinne Houart
- Medical Research Council Centre for Developmental Neurobiology, King's College London, London SE1 1UL, UK
| | - Evan Reid
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 OXY, UK
| | - Jamilé Hazan
- Sorbonne Universités, UPMC Université Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), 75005 Paris, France
| | - Coralie Fassier
- Sorbonne Universités, UPMC Université Paris 06, INSERM, CNRS, Neuroscience Paris Seine - Institut de Biologie Paris-Seine (NPS-IBPS), 75005 Paris, France
| |
Collapse
|
165
|
Paatero I, Sauteur L, Lee M, Lagendijk AK, Heutschi D, Wiesner C, Guzmán C, Bieli D, Hogan BM, Affolter M, Belting HG. Junction-based lamellipodia drive endothelial cell rearrangements in vivo via a VE-cadherin-F-actin based oscillatory cell-cell interaction. Nat Commun 2018; 9:3545. [PMID: 30171187 PMCID: PMC6119192 DOI: 10.1038/s41467-018-05851-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/26/2018] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis and vascular remodeling are driven by extensive endothelial cell movements. Here, we present in vivo evidence that endothelial cell movements are associated with oscillating lamellipodia-like structures, which emerge from cell junctions in the direction of cell movements. High-resolution time-lapse imaging of these junction-based lamellipodia (JBL) shows dynamic and distinct deployment of junctional proteins, such as F-actin, VE-cadherin and ZO1, during JBL oscillations. Upon initiation, F-actin and VE-cadherin are broadly distributed within JBL, whereas ZO1 remains at cell junctions. Subsequently, a new junction is formed at the front of the JBL, which then merges with the proximal junction. Rac1 inhibition interferes with JBL oscillations and disrupts cell elongation-similar to a truncation in ve-cadherin preventing VE-cad/F-actin interaction. Taken together, our observations suggest an oscillating ratchet-like mechanism, which is used by endothelial cells to move over each other and thus provides the physical means for cell rearrangements.
Collapse
Affiliation(s)
- Ilkka Paatero
- Department of Cell Biology, Biozentrum, University of Basel, Basel, 4056, Switzerland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Loïc Sauteur
- Department of Cell Biology, Biozentrum, University of Basel, Basel, 4056, Switzerland
| | - Minkyoung Lee
- Department of Cell Biology, Biozentrum, University of Basel, Basel, 4056, Switzerland
| | - Anne K Lagendijk
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Daniel Heutschi
- Department of Cell Biology, Biozentrum, University of Basel, Basel, 4056, Switzerland
| | - Cora Wiesner
- Department of Cell Biology, Biozentrum, University of Basel, Basel, 4056, Switzerland
| | - Camilo Guzmán
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Dimitri Bieli
- Department of Cell Biology, Biozentrum, University of Basel, Basel, 4056, Switzerland
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Markus Affolter
- Department of Cell Biology, Biozentrum, University of Basel, Basel, 4056, Switzerland.
| | - Heinz-Georg Belting
- Department of Cell Biology, Biozentrum, University of Basel, Basel, 4056, Switzerland.
| |
Collapse
|
166
|
Lancino M, Majello S, Herbert S, De Chaumont F, Tinevez JY, Olivo-Marin JC, Herbomel P, Schmidt A. Anisotropic organization of circumferential actomyosin characterizes hematopoietic stem cells emergence in the zebrafish. eLife 2018; 7:37355. [PMID: 30132756 PMCID: PMC6105311 DOI: 10.7554/elife.37355] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 07/28/2018] [Indexed: 12/15/2022] Open
Abstract
Hematopoiesis leads to the formation of blood and immune cells. Hematopoietic stem cells emerge during development, from vascular components, via a process called the endothelial-to-hematopoietic transition (EHT). Here, we reveal essential biomechanical features of the EHT, using the zebrafish embryo imaged at unprecedented spatio-temporal resolution and an algorithm to unwrap the aorta into 2D-cartography. We show that the transition involves anisotropic contraction along the antero-posterior axis, with heterogenous organization of contractile circumferential actomyosin. The biomechanics of the contraction is oscillatory, with unusually long periods in comparison to other apical constriction mechanisms described so far in morphogenesis, and is supported by the anisotropic reinforcement of junctional contacts. Finally, we show that abrogation of blood flow impairs the actin cytoskeleton, the morphodynamics of EHT cells, and the orientation of the emergence. Overall, our results underline the peculiarities of the EHT biomechanics and the influence of the mechanical forces exerted by blood flow. As humans, we have two major types of blood cell: our red blood cells transport oxygen around the body, while our white blood cells fight disease. Both types of cell come from the same stem cells, which first appear early in embryonic development. These stem cells emerge from the walls of major blood vessels, including the aorta – which carries blood from the heart. Stem cells have not yet decided which adult cell to become. Given the right signals, blood stem cells can form red blood cells or any of the different types of white blood cell. Understanding this process could allow scientists to recreate it in the laboratory, making blood stem cells that can give rise to all blood cells found in the body. But, this is not yet possible because we do not know all the conditions needed to make the cells and ensure they survive. One crucial gap in our understanding concerns the importance of blood flow. As the main blood vessel leaving the heart, the aorta experiences mechanical stress every time the heart beats. Lancino et al. wanted to find out whether this influences the development of the blood stem cells. Zebrafish embryos are transparent, making it easy to see their bodies developing under a microscope. Like humans, they also produce both red blood cells and white blood cells meaning Lancino et al. could watch the birth of blood stem cells in these embryos from a part of the aorta called the aortic floor. A new software tool unwrapped pictures of the tube-shaped blood vessel into flat, two-dimensional maps, making it possible to see how the aorta changed over time. This revealed that, as blood stem cells leave the aortic floor, they bend and contract with the direction of the blood flow. Rings of actin and myosin proteins that formed around the stem cells as they are born helped the process along, while stopping the heartbeat changed the way the blood cells emerged. Without any blood flow, the actin proteins did not assemble properly; the stem cells also emerged in the wrong direction and some of them even died. These findings show that physical forces play a role in the formation of blood stem cells. Understanding this process brings scientists a step closer to recreating the conditions for making different kinds of blood cells outside of the body.
Collapse
Affiliation(s)
- Mylene Lancino
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS, UMR 3738, Paris, France.,Sorbonne Université, UPMC Paris 06, Complexité du Vivant, Paris, France
| | - Sara Majello
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS, UMR 3738, Paris, France
| | - Sebastien Herbert
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS, UMR 3738, Paris, France.,Image Analysis Hub, UTechSPhotonic BioImaging (Imagopole), Citech, Institut Pasteur, Paris, France
| | - Fabrice De Chaumont
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France.,CNRS, UMR3691, Paris, France
| | - Jean-Yves Tinevez
- Image Analysis Hub, UTechSPhotonic BioImaging (Imagopole), Citech, Institut Pasteur, Paris, France
| | | | - Philippe Herbomel
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS, UMR 3738, Paris, France
| | - Anne Schmidt
- Department of Developmental and Stem Cell Biology, Institut Pasteur, Paris, France.,CNRS, UMR 3738, Paris, France
| |
Collapse
|
167
|
Zhang H, Zhang Q, Gao G, Wang X, Wang T, Kong Z, Wang G, Zhang C, Wang Y, Peng G. UBTOR/KIAA1024 regulates neurite outgrowth and neoplasia through mTOR signaling. PLoS Genet 2018; 14:e1007583. [PMID: 30080879 PMCID: PMC6095612 DOI: 10.1371/journal.pgen.1007583] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 08/16/2018] [Accepted: 07/23/2018] [Indexed: 12/22/2022] Open
Abstract
The mTOR signaling pathways regulate cell growth and are involved in multiple human diseases. Here, we identify UBTOR, a previously unannotated gene as a functional player in regulating cell growth and mTOR signaling. Reduction of UBTOR function in cultured hippocampal neurons and PC12 cells promotes neurite outgrowth. UBTOR depletion activates mTOR signaling and promotes cell growth, whilst UBTOR overexpression suppresses colony formation in cancer cell lines. Studies in cultured cells and zebrafish model show that UBTOR inhibits mTOR signaling by stabilizing the mTOR complex component DEPTOR, and ubtor gene disruption result in higher mTOR activity and aggravate HRAS(G12V) induced neoplasia in the zebrafish. Lastly, UBTOR depletion promotes tumor growth and mTOR signaling in a xenograft mouse model. Together, our results demonstrate how UBTOR regulates cell growth and neoplasia via mTOR signaling. Cell growth is a fundamental aspect of cell behavior in all organisms. The mTOR signaling pathways are essential for cell growth and clinically mis-regulation of the mTOR pathways are implicated in human diseases including tumor formation, obesity, epilepsy, autism and neurodegeneration. Here, we identify a novel gene, Ubtor as a functional player in regulating cell growth and mTOR signaling. Inhibiting Ubtor function promotes cell growth in neurons and cancer cells. Increasing Ubtor function reduces cancer cell growth. Functional analyses in human cells and the zebrafish model indicate Ubtor inhibits mTOR signaling by stabilizing the mTOR complex component DEPTOR, and ubtor gene disruption resulted in higher mTOR activity and aggravated cancer formation in the zebrafish. UBTOR depletion promotes tumor growth and mTOR signaling in xenograft-bearing mice. Thus our study provide evidence that Ubtor constitutes a novel negative feedback mechanism to control mTOR signaling and cell growth, and manipulations of Ubtor function may potentially be utilized to optimize mTOR signaling activities for treatments of cancers and other diseases.
Collapse
Affiliation(s)
- Hefei Zhang
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Quan Zhang
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Ge Gao
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Xinjian Wang
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Tiantian Wang
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Zhitao Kong
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Guoxiang Wang
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Cuizhen Zhang
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Yun Wang
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
| | - Gang Peng
- Institute of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, China
- * E-mail:
| |
Collapse
|
168
|
Tanaka Y, Hayashi K, Fujino N, Konno T, Tada H, Nakanishi C, Hodatsu A, Tsuda T, Nagata Y, Teramoto R, Yoshida S, Nomura A, Kawashiri MA, Yamagishi M. Functional analysis of KCNH2 gene mutations of type 2 long QT syndrome in larval zebrafish using microscopy and electrocardiography. Heart Vessels 2018; 34:159-166. [PMID: 30047011 DOI: 10.1007/s00380-018-1231-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 07/20/2018] [Indexed: 10/28/2022]
Abstract
Heterologous expression systems play a vital role in the characterization of potassium voltage-gated channel subfamily H member 2 (KCNH2) gene mutations, such as E637K which is associated with long QT syndrome type 2 (LQT2). In vivo assays using zebrafish provide a means for testing genetic variants of cardiac disease; however, limited information on the role of the E637K mutation is available from in vivo systems and their utility has yet to be fully exploited in the context of LQT2. We sought to evaluate the ability of the E637K mutant channel to restore normal repolarization in larval zebrafish with a human KCNH2 orthologue, kcnh2a-knockdown. A morpholino (MO) targeting kcnh2a was injected alone or with wild type (WT) or E637K KCNH2 cRNA into zebrafish embryos at the 1-2 cell stage. Cardiac repolarization phenotypes were screened using light microscopy and the QT interval was measured by single lead electrocardiograph (ECG) analysis at 72-h post-fertilization. In the MO alone group, 17% of zebrafish had a normal phenotype; this rate increased to 60% in the WT KCNH2 cRNA injected zebrafish and to 35% in the E637K injected zebrafish. The ECG of larval zebrafish revealed that QTc was significantly prolonged in the MO alone group compared to the control group. Co-injection of WT KCNH2 cRNA shortened the QTc interval, however, that of the E637K did not. We suggest that this in vivo cardiac assay using microscopy and ECG in larval zebrafish offers a reliable approach for risk discrimination of KCNH2 mutations.
Collapse
Affiliation(s)
- Yoshihiro Tanaka
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Kenshi Hayashi
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
| | - Noboru Fujino
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Tetsuo Konno
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hayato Tada
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Chiaki Nakanishi
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Akihiko Hodatsu
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Toyonobu Tsuda
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yoji Nagata
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Ryota Teramoto
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Shohei Yoshida
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Akihiro Nomura
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masa-Aki Kawashiri
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Masakazu Yamagishi
- Department of Cardiovascular and Internal Medicine, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| |
Collapse
|
169
|
Miller GW, Chandrasekaran V, Yaghoobi B, Lein PJ. Opportunities and challenges for using the zebrafish to study neuronal connectivity as an endpoint of developmental neurotoxicity. Neurotoxicology 2018; 67:102-111. [PMID: 29704525 PMCID: PMC6177215 DOI: 10.1016/j.neuro.2018.04.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/23/2018] [Accepted: 04/24/2018] [Indexed: 01/28/2023]
Abstract
Chemical exposures have been implicated as environmental risk factors that interact with genetic susceptibilities to influence individual risk for complex neurodevelopmental disorders, including autism spectrum disorder, schizophrenia, attention deficit hyperactivity disorder and intellectual disabilities. Altered patterns of neuronal connectivity represent a convergent mechanism of pathogenesis for these and other neurodevelopmental disorders, and growing evidence suggests that chemicals can interfere with specific signaling pathways that regulate the development of neuronal connections. There is, therefore, a growing interest in developing screening platforms to identify chemicals that alter neuronal connectivity. Cell-cell, cell-matrix interactions and systemic influences are known to be important in defining neuronal connectivity in the developing brain, thus, a systems-based model offers significant advantages over cell-based models for screening chemicals for effects on neuronal connectivity. The embryonic zebrafish represents a vertebrate model amenable to higher throughput chemical screening that has proven useful in characterizing conserved mechanisms of neurodevelopment. Moreover, the zebrafish is readily amenable to gene editing to integrate genetic susceptibilities. Although use of the zebrafish model in toxicity testing has increased in recent years, the diverse tools available for imaging structural differences in the developing zebrafish brain have not been widely applied to studies of the influence of gene by environment interactions on neuronal connectivity in the developing zebrafish brain. Here, we discuss tools available for imaging of neuronal connectivity in the developing zebrafish, review what has been published in this regard, and suggest a path forward for applying this information to developmental neurotoxicity testing.
Collapse
Affiliation(s)
- Galen W. Miller
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| | - Vidya Chandrasekaran
- Department of Biology, Saint Mary’s College of California, Moraga, CA 94575, USA
| | - Bianca Yaghoobi
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
170
|
Grajevskaja V, Camerota D, Bellipanni G, Balciuniene J, Balciunas D. Analysis of a conditional gene trap reveals that tbx5a is required for heart regeneration in zebrafish. PLoS One 2018; 13:e0197293. [PMID: 29933372 PMCID: PMC6014646 DOI: 10.1371/journal.pone.0197293] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/30/2018] [Indexed: 01/27/2023] Open
Abstract
The ability to conditionally inactivate genes is instrumental for fine genetic analysis of all biological processes, but is especially important for studies of biological events, such as regeneration, which occur late in ontogenesis or in adult life. We have constructed and tested a fully conditional gene trap vector, and used it to inactivate tbx5a in the cardiomyocytes of larval and adult zebrafish. We observe that loss of tbx5a function significantly impairs the ability of zebrafish hearts to regenerate after ventricular resection, indicating that Tbx5a plays an essential role in the transcriptional program of heart regeneration.
Collapse
Affiliation(s)
- Viktorija Grajevskaja
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States of America
- Department of Zoology, Faculty of Natural Sciences, Vilnius University, Vilnius, Lithuania
| | - Diana Camerota
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States of America
| | - Gianfranco Bellipanni
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States of America
| | - Jorune Balciuniene
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States of America
| | - Darius Balciunas
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, United States of America
| |
Collapse
|
171
|
Yang YHC, Kawakami K, Stainier DY. A new mode of pancreatic islet innervation revealed by live imaging in zebrafish. eLife 2018; 7:34519. [PMID: 29916364 PMCID: PMC6039180 DOI: 10.7554/elife.34519] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 06/18/2018] [Indexed: 12/13/2022] Open
Abstract
Pancreatic islets are innervated by autonomic and sensory nerves that influence their function. Analyzing the innervation process should provide insight into the nerve-endocrine interactions and their roles in development and disease. Here, using in vivo time-lapse imaging and genetic analyses in zebrafish, we determined the events leading to islet innervation. Comparable neural density in the absence of vasculature indicates that it is dispensable for early pancreatic innervation. Neural crest cells are in close contact with endocrine cells early in development. We find these cells give rise to neurons that extend axons toward the islet as they surprisingly migrate away. Specific ablation of these neurons partly prevents other neurons from migrating away from the islet resulting in diminished innervation. Thus, our studies establish the zebrafish as a model to interrogate mechanisms of organ innervation, and reveal a novel mode of innervation whereby neurons establish connections with their targets before migrating away.
Collapse
Affiliation(s)
- Yu Hsuan Carol Yang
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan.,Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Japan
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
172
|
In vitro evaluation of a lentiviral two-step transcriptional amplification system using GAL4FF transactivator for gene therapy applications in bone repair. Gene Ther 2018; 25:260-268. [PMID: 29907876 DOI: 10.1038/s41434-018-0024-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/02/2018] [Accepted: 05/23/2018] [Indexed: 01/20/2023]
Abstract
In this study, we developed a lentiviral two-step transcriptional amplification (TSTA) system expressing bone morphogenetic protein-2 (BMP-2) under the control of a GAL4FF transactivator to enhance gene expression and limit toxicity for bone repair applications. To this end human MSCs, isolated from bone marrow or adipose tissue, were transduced overnight with a LV-TSTA system (GAL4FF or GAL4vp16) expressing BMP-2 or GFP and evaluated in vitro for transduction efficiency, mean fluorescence intensity, cell viability, and BMP-2 production. FACS analysis of GFP-transduced MSCs confirmed successful transduction with the GAL4FF+GFP vector. Moreover, ELISA demonstrated abundant BMP-2 production by GAL4FF+BMP2-transduced human MSCs over a period of 8 weeks, with minimal cytotoxicity at all time points. Compared to GAL4vp16, GAL4FF was superior with respect to BMP production at 1, 2, 4, 6, and 8 weeks in BMSCs. In ASCs, GAL4FF was still associated with higher BMP-2 production at weeks 2-8, but this difference was not as prominent as in BMSCs. To our knowledge, this is the first report of GAL4FF-mediated BMP-2 production by human BMSCs and ASCs. Compared to the standard GAL4vp16TSTA vector, GAL4FF was associated with lower cytotoxicity and higher in vitro gene expression in both BMSCs and ASCs.
Collapse
|
173
|
Muto A, Kawakami K. Ablation of a Neuronal Population Using a Two-photon Laser and Its Assessment Using Calcium Imaging and Behavioral Recording in Zebrafish Larvae. J Vis Exp 2018. [PMID: 29912192 DOI: 10.3791/57485] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
To identify the role of a subpopulation of neurons in behavior, it is essential to test the consequences of blocking its activity in living animals. Laser ablation of neurons is an effective method for this purpose when neurons are selectively labeled with fluorescent probes. In the present study, protocols for laser ablating a subpopulation of neurons using a two-photon microscope and testing of its functional and behavioral consequences are described. In this study, prey capture behavior in zebrafish larvae is used as a study model. The pretecto-hypothalamic circuit is known to underlie this visually-driven prey catching behavior. Zebrafish pretectum were laser-ablated, and neuronal activity in the inferior lobe of the hypothalamus (ILH; the target of the pretectal projection) was examined. Prey capture behavior after pretectal ablation was also tested.
Collapse
Affiliation(s)
- Akira Muto
- Division of Molecular and Developmental Biology, National Institute of Genetics, Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies);
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies)
| |
Collapse
|
174
|
Fukui H, Miyazaki T, Chow RWY, Ishikawa H, Nakajima H, Vermot J, Mochizuki N. Hippo signaling determines the number of venous pole cells that originate from the anterior lateral plate mesoderm in zebrafish. eLife 2018; 7:29106. [PMID: 29809141 PMCID: PMC5995544 DOI: 10.7554/elife.29106] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 05/26/2018] [Indexed: 12/11/2022] Open
Abstract
The differentiation of the lateral plate mesoderm cells into heart field cells constitutes a critical step in the development of cardiac tissue and the genesis of functional cardiomyocytes. Hippo signaling controls cardiomyocyte proliferation, but the role of Hippo signaling during early cardiogenesis remains unclear. Here, we show that Hippo signaling regulates atrial cell number by specifying the developmental potential of cells within the anterior lateral plate mesoderm (ALPM), which are incorporated into the venous pole of the heart tube and ultimately into the atrium of the heart. We demonstrate that Hippo signaling acts through large tumor suppressor kinase 1/2 to modulate BMP signaling and the expression of hand2, a key transcription factor that is involved in the differentiation of atrial cardiomyocytes. Collectively, these results demonstrate that Hippo signaling defines venous pole cardiomyocyte number by modulating both the number and the identity of the ALPM cells that will populate the atrium of the heart.
Collapse
Affiliation(s)
- Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,University of Strasbourg Institute for Advanced Study (USIAS), Strasbourg, France.,Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France.,Université de Strasbourg, Illkirch, France
| | - Takahiro Miyazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Renee Wei-Yan Chow
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Hiroyuki Ishikawa
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | - Julien Vermot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France.,Centre National de la Recherche Scientifique, Illkirch, France.,Université de Strasbourg, Illkirch, France.,Institut National de la Santé et de la Recherche Médicale, Illkirch, France
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan.,AMED-Core Research for Evolutional Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| |
Collapse
|
175
|
Guerra A, Germano RF, Stone O, Arnaout R, Guenther S, Ahuja S, Uribe V, Vanhollebeke B, Stainier DY, Reischauer S. Distinct myocardial lineages break atrial symmetry during cardiogenesis in zebrafish. eLife 2018; 7:32833. [PMID: 29762122 PMCID: PMC5953537 DOI: 10.7554/elife.32833] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 04/04/2018] [Indexed: 02/06/2023] Open
Abstract
The ultimate formation of a four-chambered heart allowing the separation of the pulmonary and systemic circuits was key for the evolutionary success of tetrapods. Complex processes of cell diversification and tissue morphogenesis allow the left and right cardiac compartments to become distinct but remain poorly understood. Here, we describe an unexpected laterality in the single zebrafish atrium analogous to that of the two atria in amniotes, including mammals. This laterality appears to derive from an embryonic antero-posterior asymmetry revealed by the expression of the transcription factor gene meis2b. In adult zebrafish hearts, meis2b expression is restricted to the left side of the atrium where it controls the expression of pitx2c, a regulator of left atrial identity in mammals. Altogether, our studies suggest that the multi-chambered atrium in amniotes arose from a molecular blueprint present before the evolutionary emergence of cardiac septation and provide insights into the establishment of atrial asymmetry.
Collapse
Affiliation(s)
- Almary Guerra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Raoul Fv Germano
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Bruxelles, Belgium
| | - Oliver Stone
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Rima Arnaout
- Division of Cardiology, Department of Medicine, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, United States
| | - Stefan Guenther
- ECCPS Bioinformatics and Deep Sequencing Platform, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Suchit Ahuja
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Verónica Uribe
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Bruxelles, Belgium
| | - Didier Yr Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| |
Collapse
|
176
|
Davis JA, Koenig AL, Lubert A, Chestnut B, Liu F, Palencia Desai S, Winkler T, Pociute K, Choi K, Sumanas S. ETS transcription factor Etsrp / Etv2 is required for lymphangiogenesis and directly regulates vegfr3 / flt4 expression. Dev Biol 2018; 440:40-52. [PMID: 29753018 DOI: 10.1016/j.ydbio.2018.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 01/05/2023]
Abstract
The molecular mechanisms initiating the formation of the lymphatic system, lymphangiogenesis, are still poorly understood. Here we have identified a novel role in lymphangiogenesis for an ETS transcription factor, Etv2/Etsrp, a known regulator of embryonic vascular development. Through the use of fully validated photoactivatable morpholinos we show that inducible Etv2 inhibition in zebrafish embryos at 1 day post-fertilization (dpf) results in significant inhibition of lymphangiogenesis, while development of blood vessels is unaffected. In Etv2-inhibited embryos and larvae, the number of lymphatic progenitors is greatly reduced, the major lymphatic vessel, the thoracic duct, is absent or severely fragmented, and lymphangiogenesis-associated marker expression, including lyve1b, prox1a, and vegfr3/flt4, is strongly downregulated. We also demonstrate that lymphatic progenitors in Etv2 deficient embryos fail to respond to Vegfc signaling. Chromatin immunoprecipitation and sequencing (ChIP-Seq) studies using differentiated mouse embryonic stem (ES) cells as well as luciferase reporter studies in the ES cells and in zebrafish embryos argue that Etv2 directly binds the promoter/enhancer regions of Vegfc receptor Vegfr3/Flt4 and lymphatic marker Lyve1, and promotes their expression. Together these data support a model where Etv2 initiates lymphangiogenesis by directly promoting the expression of flt4 within the posterior cardinal vein.
Collapse
Affiliation(s)
- Jennifer A Davis
- Cancer&Blood Disease Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Andrew L Koenig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Allison Lubert
- Cancer&Blood Disease Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Brendan Chestnut
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Fang Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sharina Palencia Desai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tamara Winkler
- Cancer&Blood Disease Institute, Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Karolina Pociute
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Graduate School of Biotechnology, Kyung Hee University, Yong In, South Korea
| | - Saulius Sumanas
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
177
|
Enya S, Kawakami K, Suzuki Y, Kawaoka S. A novel zebrafish intestinal tumor model reveals a role for cyp7a1-dependent tumor-liver crosstalk in causing adverse effects on the host. Dis Model Mech 2018; 11:dmm.032383. [PMID: 29592890 PMCID: PMC6124559 DOI: 10.1242/dmm.032383] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 03/12/2018] [Indexed: 12/18/2022] Open
Abstract
The nature of host organs and genes that underlie tumor-induced physiological disruption on the host remains ill-defined. Here, we establish a novel zebrafish intestinal tumor model that is suitable for addressing this issue, and find that hepatic cyp7a1, the rate-limiting factor for synthesizing bile acids [or, in the case of zebrafish, bile alcohol (BA)], is such a host gene. Inducing krasG12D by Gal4 specifically expressed in the posterior intestine resulted in the formation of an intestinal tumor. The local intestinal tumor caused systemic detrimental effects on the host, including liver inflammation, hepatomegaly, growth defects and organismal death. Whole-organism-level gene expression analysis and metabolite measurements revealed that the intestinal tumor reduced total BA levels, possibly via altered expression of hepatic cyp7a1 Genetically overexpressing cyp7a1 in the liver restored BA synthesis and ameliorated tumor-induced liver inflammation, but not other tumor-dependent phenotypes. Thus, we found a previously unknown role of cyp7a1 as the host gene that links the intestinal tumor, hepatic cholesterol-BA metabolism and liver inflammation in tumor-bearing zebrafish larvae. Our model provides an important basis to discover host genes responsible for tumor-induced phenotypes and to uncover mechanisms underlying how tumors adversely affect host organisms.
Collapse
Affiliation(s)
- Sora Enya
- Advanced Telecommunications Research Institute International (ATR), The Thomas N. Sato BioMEC-X Laboratories, Kyoto 619-0288, Japan.,ERATO Sato Live Bio-forecasting Project, Japan Science and Technology Agency (JST), Kyoto 619-0288, Japan
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, and Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka 411-8540, Japan
| | - Yutaka Suzuki
- The University of Tokyo, Graduate School of Frontier Science, Kashiwa 277-8651, Japan
| | - Shinpei Kawaoka
- Advanced Telecommunications Research Institute International (ATR), The Thomas N. Sato BioMEC-X Laboratories, Kyoto 619-0288, Japan .,ERATO Sato Live Bio-forecasting Project, Japan Science and Technology Agency (JST), Kyoto 619-0288, Japan
| |
Collapse
|
178
|
Epidermal regulation of bone morphogenesis through the development and regeneration of osteoblasts in the zebrafish scale. Dev Biol 2018. [DOI: 10.1016/j.ydbio.2018.03.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
179
|
Lal P, Tanabe H, Suster ML, Ailani D, Kotani Y, Muto A, Itoh M, Iwasaki M, Wada H, Yaksi E, Kawakami K. Identification of a neuronal population in the telencephalon essential for fear conditioning in zebrafish. BMC Biol 2018; 16:45. [PMID: 29690872 PMCID: PMC5978991 DOI: 10.1186/s12915-018-0502-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 03/07/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fear conditioning is a form of learning essential for animal survival and used as a behavioral paradigm to study the mechanisms of learning and memory. In mammals, the amygdala plays a crucial role in fear conditioning. In teleost, the medial zone of the dorsal telencephalon (Dm) has been postulated to be a homolog of the mammalian amygdala by anatomical and ablation studies, showing a role in conditioned avoidance response. However, the neuronal populations required for a conditioned avoidance response via the Dm have not been functionally or genetically defined. RESULTS We aimed to identify the neuronal population essential for fear conditioning through a genetic approach in zebrafish. First, we performed large-scale gene trap and enhancer trap screens, and created transgenic fish lines that expressed Gal4FF, an engineered version of the Gal4 transcription activator, in specific regions in the brain. We then crossed these Gal4FF-expressing fish with the effector line carrying the botulinum neurotoxin gene downstream of the Gal4 binding sequence UAS, and analyzed the double transgenic fish for active avoidance fear conditioning. We identified 16 transgenic lines with Gal4FF expression in various brain areas showing reduced performance in avoidance responses. Two of them had Gal4 expression in populations of neurons located in subregions of the Dm, which we named 120A-Dm neurons. Inhibition of the 120A-Dm neurons also caused reduced performance in Pavlovian fear conditioning. The 120A-Dm neurons were mostly glutamatergic and had projections to other brain regions, including the hypothalamus and ventral telencephalon. CONCLUSIONS Herein, we identified a subpopulation of neurons in the zebrafish Dm essential for fear conditioning. We propose that these are functional equivalents of neurons in the mammalian pallial amygdala, mediating the conditioned stimulus-unconditioned stimulus association. Thus, the study establishes a basis for understanding the evolutionary conservation and diversification of functional neural circuits mediating fear conditioning in vertebrates.
Collapse
Affiliation(s)
- Pradeep Lal
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
- Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka, 411-8540 Japan
- Kavli Institute for Systems Neuroscience and Centre for the Biology of Memory, Norwegian Brain Centre, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Hideyuki Tanabe
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
| | - Maximiliano L. Suster
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
- Present address: Visual Interaction GmbH, Warthestrasse 21, 14513 Teltow, Germany
| | - Deepak Ailani
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
| | - Yuri Kotani
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
| | - Akira Muto
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
| | - Mari Itoh
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
| | - Miki Iwasaki
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
- Present address: College of Liberal Arts and Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373 Japan
| | - Hironori Wada
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
- Present address: College of Liberal Arts and Sciences, Kitasato University, Sagamihara, Kanagawa 252-0373 Japan
| | - Emre Yaksi
- Kavli Institute for Systems Neuroscience and Centre for the Biology of Memory, Norwegian Brain Centre, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Shizuoka, 411-8540 Japan
- Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, Shizuoka, 411-8540 Japan
| |
Collapse
|
180
|
Ishikawa K, Kobayashi Y, Wakabayashi Y, Watanabe S, Semba K. A highly sensitive trap vector system for isolating reporter cells and identification of responsive genes. Biol Methods Protoc 2018; 3:bpy003. [PMID: 32161797 PMCID: PMC6994077 DOI: 10.1093/biomethods/bpy003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/22/2018] [Accepted: 04/18/2018] [Indexed: 01/03/2023] Open
Abstract
We devised a versatile vector system for efficient isolation of reporter cells responding to a certain condition of interest. This system combines nontoxic GAL4-UAS and piggyBac transposon systems, allowing application to mammalian cells and improved expression of a fluorescent reporter protein for cell sorting. Case studies under conditions of c-MYC gene induction or endoplasmic reticulum (ER) stress with thapsigargin on mouse or human cell lines confirmed easy and efficient isolation of responsive reporter cells. Sequence analyses of the integrated loci of the thapsigargin-responsive clones identified responsive genes including BiP and OSBPL9. OSBPL9 is a novel ER stress-responsive gene and we confirmed that endogenous mRNA expression of OSBPL9 is upregulated by thapsigargin, and is repressed by IRE1α inhibitors, 4μ8C and toyocamycin, but not significantly by a PERK inhibitor, GSK2656157. These results demonstrate that this approach can be used to discover novel genes regulated by any stimuli without the need for microarray analysis, and that it can concomitantly produce reporter cells without identification of stimuli-responsive promoter/enhancer elements. Therefore, this system has a variety of benefits for basic and clinical research.
Collapse
Affiliation(s)
- Kosuke Ishikawa
- Japan Biological Informatics Consortium (JBiC), 2-45 Aomi, Koto-ku, Tokyo 135-8073, Japan
| | - Yuta Kobayashi
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Yutaro Wakabayashi
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Shinya Watanabe
- Translational Research Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu-cho, Shinjuku-ku, Tokyo 162-8480, Japan.,Translational Research Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan
| |
Collapse
|
181
|
Optical interrogation of neuronal circuitry in zebrafish using genetically encoded voltage indicators. Sci Rep 2018; 8:6048. [PMID: 29662090 PMCID: PMC5902623 DOI: 10.1038/s41598-018-23906-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 03/20/2018] [Indexed: 11/21/2022] Open
Abstract
Optical measurement of membrane potentials enables fast, direct and simultaneous detection of membrane potentials from a population of neurons, providing a desirable approach for functional analysis of neuronal circuits. Here, we applied recently developed genetically encoded voltage indicators, ASAP1 (Accelerated Sensor of Action Potentials 1) and QuasAr2 (Quality superior to Arch 2), to zebrafish, an ideal model system for studying neurogenesis. To achieve this, we established transgenic lines which express the voltage sensors, and showed that ASAP1 is expressed in zebrafish neurons. To examine whether neuronal activity could be detected by ASAP1, we performed whole-cerebellum imaging, showing that depolarization was detected widely in the cerebellum and optic tectum upon electrical stimulation. Spontaneous activity in the spinal cord was also detected by ASAP1 imaging at single-cell resolution as well as at the neuronal population level. These responses mostly disappeared following treatment with tetrodotoxin, indicating that ASAP1 enabled optical measurement of neuronal activity in the zebrafish brain. Combining this method with other approaches, such as optogenetics and behavioural analysis may facilitate a deeper understanding of the functional organization of brain circuitry and its development.
Collapse
|
182
|
Integrative whole-brain neuroscience in larval zebrafish. Curr Opin Neurobiol 2018; 50:136-145. [PMID: 29486425 DOI: 10.1016/j.conb.2018.02.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/23/2018] [Accepted: 02/04/2018] [Indexed: 11/22/2022]
Abstract
Due to their small size and transparency, zebrafish larvae are amenable to a range of fluorescence microscopy techniques. With the development of sensitive genetically encoded calcium indicators, this has extended to the whole-brain imaging of neural activity with cellular resolution. This technique has been used to study brain-wide population dynamics accompanying sensory processing and sensorimotor transformations, and has spurred the development of innovative closed-loop behavioral paradigms in which stimulus-response relationships can be studied. More recently, microscopes have been developed that allow whole-brain calcium imaging in freely swimming and behaving larvae. In this review, we highlight the technologies underlying whole-brain functional imaging in zebrafish, provide examples of the sensory and motor processes that have been studied with this technique, and discuss the need to merge data from whole-brain functional imaging studies with neurochemical and anatomical information to develop holistic models of functional neural circuits.
Collapse
|
183
|
Vanlandewijck M, He L, Mäe MA, Andrae J, Ando K, Del Gaudio F, Nahar K, Lebouvier T, Laviña B, Gouveia L, Sun Y, Raschperger E, Räsänen M, Zarb Y, Mochizuki N, Keller A, Lendahl U, Betsholtz C. A molecular atlas of cell types and zonation in the brain vasculature. Nature 2018; 554:475-480. [PMID: 29443965 DOI: 10.1038/nature25739] [Citation(s) in RCA: 1258] [Impact Index Per Article: 179.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 01/10/2018] [Indexed: 01/04/2023]
Abstract
Cerebrovascular disease is the third most common cause of death in developed countries, but our understanding of the cells that compose the cerebral vasculature is limited. Here, using vascular single-cell transcriptomics, we provide molecular definitions for the principal types of blood vascular and vessel-associated cells in the adult mouse brain. We uncover the transcriptional basis of the gradual phenotypic change (zonation) along the arteriovenous axis and reveal unexpected cell type differences: a seamless continuum for endothelial cells versus a punctuated continuum for mural cells. We also provide insight into pericyte organotypicity and define a population of perivascular fibroblast-like cells that are present on all vessel types except capillaries. Our work illustrates the power of single-cell transcriptomics to decode the higher organizational principles of a tissue and may provide the initial chapter in a molecular encyclopaedia of the mammalian vasculature.
Collapse
Affiliation(s)
- Michael Vanlandewijck
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Blickagången 6, SE-141 57 Huddinge, Sweden.,Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Liqun He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin 300052, China
| | - Maarja Andaloussi Mäe
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Johanna Andrae
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Koji Ando
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Francesca Del Gaudio
- Department of Cell and Molecular Biology, Karolinska Institutet, Von Eulers väg 3, SE-171 77 Stockholm, Sweden
| | - Khayrun Nahar
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Thibaud Lebouvier
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden.,Inserm U1171, University of Lille, CHU, Memory Center, Distalz, F-59000 Lille, France
| | - Bàrbara Laviña
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Leonor Gouveia
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| | - Ying Sun
- Department of Bioinformatics, Zhongyuan Union Genetic Technology Co., Ltd., No.45, the 9th East Road, Tianjin Airport Economic Area, Tianjin 300304, China
| | - Elisabeth Raschperger
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Blickagången 6, SE-141 57 Huddinge, Sweden
| | - Markus Räsänen
- Wihuri Research Institute and Translational Cancer Biology Program, Biomedicum Helsinki, University of Helsinki, Haartmaninkatu 8, P.O. Box 63, FI-00014 Helsinki, Finland
| | - Yvette Zarb
- Division of Neurosurgery, Zürich University Hospital, Zürich University, Zürich, CH-8091, Switzerland
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.,AMED-CREST, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Annika Keller
- Division of Neurosurgery, Zürich University Hospital, Zürich University, Zürich, CH-8091, Switzerland
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Von Eulers väg 3, SE-171 77 Stockholm, Sweden
| | - Christer Betsholtz
- Karolinska Institutet/AstraZeneca Integrated Cardio Metabolic Centre (KI/AZ ICMC), Blickagången 6, SE-141 57 Huddinge, Sweden.,Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, SE-751 85 Uppsala, Sweden
| |
Collapse
|
184
|
Burkhard SB, Bakkers J. Spatially resolved RNA-sequencing of the embryonic heart identifies a role for Wnt/β-catenin signaling in autonomic control of heart rate. eLife 2018; 7:31515. [PMID: 29400650 PMCID: PMC5815850 DOI: 10.7554/elife.31515] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 02/03/2018] [Indexed: 11/13/2022] Open
Abstract
Development of specialized cells and structures in the heart is regulated by spatially -restricted molecular pathways. Disruptions in these pathways can cause severe congenital cardiac malformations or functional defects. To better understand these pathways and how they regulate cardiac development we used tomo-seq, combining high-throughput RNA-sequencing with tissue-sectioning, to establish a genome-wide expression dataset with high spatial resolution for the developing zebrafish heart. Analysis of the dataset revealed over 1100 genes differentially expressed in sub-compartments. Pacemaker cells in the sinoatrial region induce heart contractions, but little is known about the mechanisms underlying their development. Using our transcriptome map, we identified spatially restricted Wnt/β-catenin signaling activity in pacemaker cells, which was controlled by Islet-1 activity. Moreover, Wnt/β-catenin signaling controls heart rate by regulating pacemaker cellular response to parasympathetic stimuli. Thus, this high-resolution transcriptome map incorporating all cell types in the embryonic heart can expose spatially restricted molecular pathways critical for specific cardiac functions.
Collapse
Affiliation(s)
- Silja Barbara Burkhard
- Hubrecht Institute-KNAW, Utrecht, Netherlands.,University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW, Utrecht, Netherlands.,University Medical Center Utrecht, Utrecht, Netherlands.,Department of Medical Physiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
185
|
Nikaido M, Izumi S, Ohnuki H, Takigawa Y, Yamasu K, Hatta K. Early development of the enteric nervous system visualized by using a new transgenic zebrafish line harboring a regulatory region for choline acetyltransferase a (chata) gene. Gene Expr Patterns 2018; 28:12-21. [PMID: 29413438 DOI: 10.1016/j.gep.2018.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
The enteric nervous system (ENS) is the largest part of the peripheral nervous system in vertebrates. Toward the visualization of the development of the vertebrate ENS, we report our creation of a new transgenic line, Tg(chata:GGFF2) which has a 1.5-kb upstream region of the zebrafish choline acetyltransferase a (chata) gene followed by modified green fluorescent protein (gfp). During development, GFP + cells were detected in the gut by 60 h post-fertilization (hpf). In the gut of 6- and 12-days post-fertilization (dpf) larvae, an average of 92% of the GFP + cells were positive for the neuronal marker HuC/D, suggesting that GFP marks enteric neurons in this transgenic line. We also observed that 66% of the GFP + cells were choline acetyltransferase (ChAT)-immunopositive at 1.5 months. Thus, GFP is expressed at the larval stages at which ChAT protein expression is not yet detected by immunostaining. We studied the spatiotemporal pattern of neural differentiation in the ENS by live-imaging of this transgenic line. We observed that GFP + or gfp + cells initially formed a pair of bilateral rows at 60 hpf or 53 hpf, respectively, in the migrating enteric neural crest cells. Most of the GFP + cells did not migrate, and most of the new GFP + cells were added to fill the space among the previously formed GFP + cells. GFP expression reached the anus by 72 hpf. New GFP + cells then also appeared in the dorsal and ventral sides of the initial GFP + rows, resulting in their distribution on the entire gut by 4 dpf. A small number of new GFP + cells were found to move among older GFP + cells just before the cells stopped migration, suggesting that the moving GFP + cells may represent neural precursor cells searching for a place for the final differentiation. Our data suggest that the Tg(chata:GGFF2) line could serve as a useful tool for studies of enteric neural differentiation and cell behavior.
Collapse
Affiliation(s)
- Masataka Nikaido
- University of Hyogo, Koto 3-2-1, Kamigori-cho, Ako-gun, Hyogo 678-1297, Japan
| | - Saki Izumi
- University of Hyogo, Koto 3-2-1, Kamigori-cho, Ako-gun, Hyogo 678-1297, Japan
| | - Honoka Ohnuki
- Saitama University, Shimo-Okubo 255, Saitama City, Saitama 338-8570, Japan
| | - Yuki Takigawa
- University of Hyogo, Koto 3-2-1, Kamigori-cho, Ako-gun, Hyogo 678-1297, Japan
| | - Kyo Yamasu
- Saitama University, Shimo-Okubo 255, Saitama City, Saitama 338-8570, Japan
| | - Kohei Hatta
- University of Hyogo, Koto 3-2-1, Kamigori-cho, Ako-gun, Hyogo 678-1297, Japan.
| |
Collapse
|
186
|
Shibata E, Ando K, Murase E, Kawakami A. Heterogeneous fates and dynamic rearrangement of regenerative epidermis-derived cells during zebrafish fin regeneration. Development 2018; 145:dev.162016. [DOI: 10.1242/dev.162016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/20/2018] [Indexed: 01/14/2023]
Abstract
The regenerative epidermis (RE) is a specialized tissue that plays an essential role in tissue regeneration. However, the fate of the RE during and after regeneration is unknown. In this study, we performed Cre-loxP-mediated cell fate tracking and revealed the fates of major population of regenerative epidermis cells that express fibronectin 1b (fn1b) during zebrafish fin regeneration. Our study showed that these RE cells are mainly recruited from the inter-ray epidermis, and that they follow heterogeneous cell fates. Early recruited cells contribute to initial wound healing and soon disappear by apoptosis, while the later recruited cells contribute to the regenerated epidermis. Intriguingly, many of these cells were also expelled from the regenerated tissue by a dynamic caudal movement of the epidermis over time, and in turn the loss of epidermal cells was replenished by a global self-replication of basal and suprabasal cells in fin. De-differentiation of non-basal epidermal cells into the basal epidermal cells did not occur during regeneration. Overall, our study revealed heterogeneous fates of RE cells and a dynamic rearrangement of the epidermis during and after regeneration.
Collapse
Affiliation(s)
- Eri Shibata
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | - Kazunori Ando
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | - Emiko Murase
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| | - Atsushi Kawakami
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
187
|
Okuda KS, Baek S, Hogan BM. Visualization and Tools for Analysis of Zebrafish Lymphatic Development. Methods Mol Biol 2018; 1846:55-70. [PMID: 30242752 DOI: 10.1007/978-1-4939-8712-2_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The accessibility and optical transparency of the zebrafish embryo offers a unique platform for live-imaging of developmental lymphangiogenesis. Transgenic lines labelling lymphatic progenitors and vessels enable researchers to visualize cellular processes and ask how they contribute to lymphatic development in genetic models. Furthermore, validated immunofluorescence staining for key signaling and cell fate markers (phosphorylated Erk and Prox1) allow single cell resolution studies of lymphatic differentiation. Here, we describe in detail how zebrafish embryos and larvae can be mounted for high resolution, staged imaging of lymphatic networks, how lymphangiogenesis can be reliably quantified and how immunofluorescence can reveal lymphatic signaling and differentiation. These methods offer researchers the opportunity to experimentally dissect developmental lymphangiogenesis with outstanding resolution.
Collapse
Affiliation(s)
- Kazuhide S Okuda
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Sungmin Baek
- Stowers Institute for Medical Research, Kansas city, MO, USA.,Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Benjamin M Hogan
- Division of Genomics of Development and Disease, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
188
|
Nagata Y, Yamagishi M, Konno T, Nakanishi C, Asano Y, Ito S, Nakajima Y, Seguchi O, Fujino N, Kawashiri MA, Takashima S, Kitakaze M, Hayashi K. Heat Failure Phenotypes Induced by Knockdown of DAPIT in Zebrafish: A New Insight into Mechanism of Dilated Cardiomyopathy. Sci Rep 2017; 7:17417. [PMID: 29234032 PMCID: PMC5727169 DOI: 10.1038/s41598-017-17572-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/28/2017] [Indexed: 11/18/2022] Open
Abstract
The pathogenesis of heart failure associated with dilated cardiomyopathy (DCM) may result in part from adenosine triphosphate (ATP) dysregulation in the myocardium. Under these conditions, diabetes-associated protein in insulin-sensitive tissue (DAPIT), which is encoded by the upregulated during skeletal muscle growth 5 (USMG5) gene, plays a crucial role in energy production by mitochondrial ATP synthase. To determine whether USMG5 is related to the development of heart failure, we performed clinical and experimental studies. Microarray analysis showed that the expression levels of USMG5 were positively correlated with those of natriuretic peptide precursor A in the human failed myocardium. When endogenous z-usmg5 in zebrafish was disrupted using morpholino (MO) oligonucleotides, the pericardial sac and atrial areas were larger and ventricular fractional shortening was reduced compared to in the control MO group. The expression levels of natriuretic peptides were upregulated in the z-usmg5 MO group compared to in controls. Further, microarray analysis revealed that genes in the calcium signalling pathway were downregulated in the z-usmg5 MO group. These results demonstrate that DAPIT plays a crucial role in the development of heart failure associated with DCM and thus may be a therapeutic target for heart failure.
Collapse
Affiliation(s)
- Yoji Nagata
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Masakazu Yamagishi
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan.
| | - Tetsuo Konno
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Chiaki Nakanishi
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Yoshihiro Asano
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Shin Ito
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Yuri Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Osamu Seguchi
- Department of Transplantation, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Noboru Fujino
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Masa-Aki Kawashiri
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masafumi Kitakaze
- Department of Clinical Research and Development, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Kenshi Hayashi
- Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| |
Collapse
|
189
|
Pathak GP, Spiltoir JI, Höglund C, Polstein LR, Heine-Koskinen S, Gersbach CA, Rossi J, Tucker CL. Bidirectional approaches for optogenetic regulation of gene expression in mammalian cells using Arabidopsis cryptochrome 2. Nucleic Acids Res 2017; 45:e167. [PMID: 28431041 PMCID: PMC5714224 DOI: 10.1093/nar/gkx260] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 04/17/2017] [Indexed: 11/13/2022] Open
Abstract
Optogenetic tools allow regulation of cellular processes with light, which can be delivered with spatiotemporal resolution. In previous work, we used cryptochrome 2 (CRY2) and CIB1, Arabidopsis proteins that interact upon light illumination, to regulate transcription with light in yeast. While adopting this approach to regulate transcription in mammalian cells, we observed light-dependent redistribution and clearing of CRY2-tethered proteins within the nucleus. The nuclear clearing phenotype was dependent on the presence of a dimerization domain contained within the CRY2-fused transcriptional activators. We used this knowledge to develop two different approaches to regulate cellular protein levels with light: a system using CRY2 and CIB1 to induce protein expression with light through stimulation of transcription, and a system using CRY2 and a LOV-fused degron to simultaneously block transcription and deplete protein levels with light. These tools will allow precise, bi-directional control of gene expression in a variety of cells and model systems.
Collapse
Affiliation(s)
- Gopal P Pathak
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jessica I Spiltoir
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Camilla Höglund
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Lauren R Polstein
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Sari Heine-Koskinen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Charles A Gersbach
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA
| | - Jari Rossi
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Chandra L Tucker
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| |
Collapse
|
190
|
Iwata T, Niimura Y, Kobayashi C, Shirakawa D, Suzuki H, Enomoto T, Touhara K, Yoshihara Y, Hirota J. A long-range cis-regulatory element for class I odorant receptor genes. Nat Commun 2017; 8:885. [PMID: 29026079 PMCID: PMC5638857 DOI: 10.1038/s41467-017-00870-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/28/2017] [Indexed: 11/09/2022] Open
Abstract
Individual olfactory sensory neurons express a single odorant receptor gene from either class I genes residing in a single cluster on a single chromosome or class II genes spread over multiple clusters on multiple chromosomes. Here, we identify an enhancer element for mouse class I genes, the J element, that is conserved through mammalian species from the platypus to humans. The J element regulates most class I genes expression by exerting an effect over ~ 3 megabases within the whole cluster. Deletion of the trans J element increases the expression frequencies of class I genes from the intact J allele, indicating that the allelic exclusion of class I genes depends on the activity of the J element. Our data reveal a long-range cis-regulatory element that governs the singular class I gene expression and has been phylogenetically preserved to retain a single cluster organization of class I genes in mammals. “Each olfactory sensory neuron expresses a single odorant receptor gene from either class I or class II genes. Here, the authors identify an enhancer for mouse class I genes, that is highly conserved, and regulates most class I genes expression by acting over ~ 3 megabases within the whole cluster.”
Collapse
Affiliation(s)
- Tetsuo Iwata
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Yoshihito Niimura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan.,ERATO Touhara Chemosensory Signal Project, The Japan Science and Technology Agency, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Chizuru Kobayashi
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Daichi Shirakawa
- Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Hikoyu Suzuki
- Nihon BioData Corporation, 3-2-1 Sakado, Takatsu-ku, Kawasaki, 213-0012, Japan
| | - Takayuki Enomoto
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501, Japan
| | - Kazushige Touhara
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan.,ERATO Touhara Chemosensory Signal Project, The Japan Science and Technology Agency, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Yoshihiro Yoshihara
- ERATO Touhara Chemosensory Signal Project, The Japan Science and Technology Agency, The University of Tokyo, Tokyo, 113-8657, Japan.,RIKEN Brain Science Institute, Saitama, 351-0198, Japan
| | - Junji Hirota
- Center for Biological Resources and Informatics, Tokyo Institute of Technology, Yokohama, 226-8501, Japan. .,Department of Life Science and Technology, Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan.
| |
Collapse
|
191
|
Kawakami K, Largaespada DA, Ivics Z. Transposons As Tools for Functional Genomics in Vertebrate Models. Trends Genet 2017; 33:784-801. [PMID: 28888423 DOI: 10.1016/j.tig.2017.07.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/14/2017] [Accepted: 07/18/2017] [Indexed: 02/06/2023]
Abstract
Genetic tools and mutagenesis strategies based on transposable elements are currently under development with a vision to link primary DNA sequence information to gene functions in vertebrate models. By virtue of their inherent capacity to insert into DNA, transposons can be developed into powerful tools for chromosomal manipulations. Transposon-based forward mutagenesis screens have numerous advantages including high throughput, easy identification of mutated alleles, and providing insight into genetic networks and pathways based on phenotypes. For example, the Sleeping Beauty transposon has become highly instrumental to induce tumors in experimental animals in a tissue-specific manner with the aim of uncovering the genetic basis of diverse cancers. Here, we describe a battery of mutagenic cassettes that can be applied in conjunction with transposon vectors to mutagenize genes, and highlight versatile experimental strategies for the generation of engineered chromosomes for loss-of-function as well as gain-of-function mutagenesis for functional gene annotation in vertebrate models, including zebrafish, mice, and rats.
Collapse
Affiliation(s)
- Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan; These authors contributed equally to this work
| | - David A Largaespada
- Department of Genetics, Cell Biology and Development, University of Minnesota, MN, USA; These authors contributed equally to this work
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany; These authors contributed equally to this work..
| |
Collapse
|
192
|
Pestel J, Ramadass R, Gauvrit S, Helker C, Herzog W, Stainier DYR. Real-time 3D visualization of cellular rearrangements during cardiac valve formation. Development 2017; 143:2217-27. [PMID: 27302398 DOI: 10.1242/dev.133272] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/26/2016] [Indexed: 12/30/2022]
Abstract
During cardiac valve development, the single-layered endocardial sheet at the atrioventricular canal (AVC) is remodeled into multilayered immature valve leaflets. Most of our knowledge about this process comes from examining fixed samples that do not allow a real-time appreciation of the intricacies of valve formation. Here, we exploit non-invasive in vivo imaging techniques to identify the dynamic cell behaviors that lead to the formation of the immature valve leaflets. We find that in zebrafish, the valve leaflets consist of two sets of endocardial cells at the luminal and abluminal side, which we refer to as luminal cells (LCs) and abluminal cells (ALCs), respectively. By analyzing cellular rearrangements during valve formation, we observed that the LCs and ALCs originate from the atrium and ventricle, respectively. Furthermore, we utilized Wnt/β-catenin and Notch signaling reporter lines to distinguish between the LCs and ALCs, and also found that cardiac contractility and/or blood flow is necessary for the endocardial expression of these signaling reporters. Thus, our 3D analyses of cardiac valve formation in zebrafish provide fundamental insights into the cellular rearrangements underlying this process.
Collapse
Affiliation(s)
- Jenny Pestel
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Radhan Ramadass
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Sebastien Gauvrit
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| | - Christian Helker
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany University of Muenster, Muenster 48149, Germany
| | - Wiebke Herzog
- University of Muenster, Muenster 48149, Germany Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Muenster 48149, Germany
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim 61231, Germany
| |
Collapse
|
193
|
Kirchberger S, Sturtzel C, Pascoal S, Distel M. Quo natas, Danio? -Recent Progress in Modeling Cancer in Zebrafish. Front Oncol 2017; 7:186. [PMID: 28894696 PMCID: PMC5581328 DOI: 10.3389/fonc.2017.00186] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 08/09/2017] [Indexed: 12/30/2022] Open
Abstract
Over the last decade, zebrafish has proven to be a powerful model in cancer research. Zebrafish form tumors that histologically and genetically resemble human cancers. The live imaging and cost-effective compound screening possible with zebrafish especially complement classic mouse cancer models. Here, we report recent progress in the field, including genetically engineered zebrafish cancer models, xenotransplantation of human cancer cells into zebrafish, promising approaches toward live investigation of the tumor microenvironment, and identification of therapeutic strategies by performing compound screens on zebrafish cancer models. Given the recent advances in genome editing, personalized zebrafish cancer models are now a realistic possibility. In addition, ongoing automation will soon allow high-throughput compound screening using zebrafish cancer models to be part of preclinical precision medicine approaches.
Collapse
Affiliation(s)
- Stefanie Kirchberger
- St. Anna Kinderkrebsforschung, Children's Cancer Research Institute, Innovative Cancer Models, Vienna, Austria
| | - Caterina Sturtzel
- St. Anna Kinderkrebsforschung, Children's Cancer Research Institute, Innovative Cancer Models, Vienna, Austria
| | - Susana Pascoal
- St. Anna Kinderkrebsforschung, Children's Cancer Research Institute, Innovative Cancer Models, Vienna, Austria
| | - Martin Distel
- St. Anna Kinderkrebsforschung, Children's Cancer Research Institute, Innovative Cancer Models, Vienna, Austria
| |
Collapse
|
194
|
Hasan SS, Tsaryk R, Lange M, Wisniewski L, Moore JC, Lawson ND, Wojciechowska K, Schnittler H, Siekmann AF. Endothelial Notch signalling limits angiogenesis via control of artery formation. Nat Cell Biol 2017; 19:928-940. [PMID: 28714969 PMCID: PMC5534340 DOI: 10.1038/ncb3574] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/15/2017] [Indexed: 01/01/2023]
Abstract
Angiogenic sprouting needs to be tightly controlled. It has been suggested that the Notch ligand dll4 expressed in leading tip cells restricts angiogenesis by activating Notch signalling in trailing stalk cells. Here, we show using live imaging in zebrafish that activation of Notch signalling is rather required in tip cells. Notch activation initially triggers expression of the chemokine receptor cxcr4a. This allows for proper tip cell migration and connection to the pre-existing arterial circulation, ultimately establishing functional arterial-venous blood flow patterns. Subsequently, Notch signalling reduces cxcr4a expression, thereby preventing excessive blood vessel growth. Finally, we find that Notch signalling is dispensable for limiting blood vessel growth during venous plexus formation that does not generate arteries. Together, these findings link the role of Notch signalling in limiting angiogenesis to its role during artery formation and provide a framework for our understanding of the mechanisms underlying blood vessel network expansion and maturation.
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Arteries/cytology
- Arteries/metabolism
- Cell Movement
- Cells, Cultured
- Endothelial Cells/metabolism
- Gene Expression Regulation, Developmental
- Genotype
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Microscopy, Fluorescence
- Microscopy, Video
- Neovascularization, Physiologic
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Phenotype
- Receptor, Notch1/genetics
- Receptor, Notch1/metabolism
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Signal Transduction
- Time Factors
- Time-Lapse Imaging
- Transfection
- Zebrafish/genetics
- Zebrafish/metabolism
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Sana S. Hasan
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, D-48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Muenster, Muenster, Germany
| | - Roman Tsaryk
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, D-48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Muenster, Muenster, Germany
| | - Martin Lange
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, D-48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Muenster, Muenster, Germany
| | - Laura Wisniewski
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, D-48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Muenster, Muenster, Germany
| | - John C. Moore
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605
| | - Nathan D. Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605
| | | | - Hans Schnittler
- Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Muenster, Muenster, Germany
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149 Münster, Germany
| | - Arndt F. Siekmann
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, D-48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Muenster, Muenster, Germany
| |
Collapse
|
195
|
Förster D, Arnold-Ammer I, Laurell E, Barker AJ, Fernandes AM, Finger-Baier K, Filosa A, Helmbrecht TO, Kölsch Y, Kühn E, Robles E, Slanchev K, Thiele TR, Baier H, Kubo F. Genetic targeting and anatomical registration of neuronal populations in the zebrafish brain with a new set of BAC transgenic tools. Sci Rep 2017; 7:5230. [PMID: 28701772 PMCID: PMC5507991 DOI: 10.1038/s41598-017-04657-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 05/19/2017] [Indexed: 11/20/2022] Open
Abstract
Genetic access to small, reproducible sets of neurons is key to an understanding of the functional wiring of the brain. Here we report the generation of a new Gal4- and Cre-driver resource for zebrafish neurobiology. Candidate genes, including cell type-specific transcription factors, neurotransmitter-synthesizing enzymes and neuropeptides, were selected according to their expression patterns in small and unique subsets of neurons from diverse brain regions. BAC recombineering, followed by Tol2 transgenesis, was used to generate driver lines that label neuronal populations in patterns that, to a large but variable extent, recapitulate the endogenous gene expression. We used image registration to characterize, compare, and digitally superimpose the labeling patterns from our newly generated transgenic lines. This analysis revealed highly restricted and mutually exclusive tissue distributions, with striking resolution of layered brain regions such as the tectum or the rhombencephalon. We further show that a combination of Gal4 and Cre transgenes allows intersectional expression of a fluorescent reporter in regions where the expression of the two drivers overlaps. Taken together, our study offers new tools for functional studies of specific neural circuits in zebrafish.
Collapse
Affiliation(s)
- Dominique Förster
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Irene Arnold-Ammer
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Eva Laurell
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Alison J Barker
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - António M Fernandes
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Karin Finger-Baier
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Alessandro Filosa
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany.,Max-Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Thomas O Helmbrecht
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Yvonne Kölsch
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Enrico Kühn
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Estuardo Robles
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany.,Department of Biological Sciences, Purdue University, West Lafayette, USA
| | - Krasimir Slanchev
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany
| | - Tod R Thiele
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany.,Department of Biological Sciences, University of Toronto Scarborough, Toronto, Canada
| | - Herwig Baier
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany.
| | - Fumi Kubo
- Max Planck Institute of Neurobiology, Department Genes - Circuits - Behavior, Am Klopferspitz 18, D-82152, Martinsried, Germany.
| |
Collapse
|
196
|
Abstract
The zebrafish (Danio rerio) possesses a vertebrate-type retina that is extraordinarily conserved in evolution. This well-organized and anatomically easily accessible part of the central nervous system has been widely investigated in zebrafish, promoting general understanding of retinal development, morphology, function and associated diseases. Over the recent years, genome and protein engineering as well as imaging techniques have experienced revolutionary advances and innovations, creating new possibilities and methods to study zebrafish development and function. In this review, we focus on some of these emerging technologies and how they may impact retinal research in the future. We place an emphasis on genetic techniques, such as transgenic approaches and the revolutionizing new possibilities in genome editing.
Collapse
Affiliation(s)
- Stephanie Niklaus
- a Institute of Molecular Life Sciences , University of Zurich , Zurich , Switzerland.,b Life Science Zurich Graduate Program - Neuroscience , Zurich , Switzerland
| | - Stephan C F Neuhauss
- a Institute of Molecular Life Sciences , University of Zurich , Zurich , Switzerland
| |
Collapse
|
197
|
Gut P, Reischauer S, Stainier DYR, Arnaout R. LITTLE FISH, BIG DATA: ZEBRAFISH AS A MODEL FOR CARDIOVASCULAR AND METABOLIC DISEASE. Physiol Rev 2017; 97:889-938. [PMID: 28468832 PMCID: PMC5817164 DOI: 10.1152/physrev.00038.2016] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/17/2022] Open
Abstract
The burden of cardiovascular and metabolic diseases worldwide is staggering. The emergence of systems approaches in biology promises new therapies, faster and cheaper diagnostics, and personalized medicine. However, a profound understanding of pathogenic mechanisms at the cellular and molecular levels remains a fundamental requirement for discovery and therapeutics. Animal models of human disease are cornerstones of drug discovery as they allow identification of novel pharmacological targets by linking gene function with pathogenesis. The zebrafish model has been used for decades to study development and pathophysiology. More than ever, the specific strengths of the zebrafish model make it a prime partner in an age of discovery transformed by big-data approaches to genomics and disease. Zebrafish share a largely conserved physiology and anatomy with mammals. They allow a wide range of genetic manipulations, including the latest genome engineering approaches. They can be bred and studied with remarkable speed, enabling a range of large-scale phenotypic screens. Finally, zebrafish demonstrate an impressive regenerative capacity scientists hope to unlock in humans. Here, we provide a comprehensive guide on applications of zebrafish to investigate cardiovascular and metabolic diseases. We delineate advantages and limitations of zebrafish models of human disease and summarize their most significant contributions to understanding disease progression to date.
Collapse
Affiliation(s)
- Philipp Gut
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Sven Reischauer
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Didier Y R Stainier
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Rima Arnaout
- Nestlé Institute of Health Sciences, EPFL Innovation Park, Lausanne, Switzerland; Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany; and Cardiovascular Research Institute and Division of Cardiology, Department of Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
198
|
Nakajima H, Yamamoto K, Agarwala S, Terai K, Fukui H, Fukuhara S, Ando K, Miyazaki T, Yokota Y, Schmelzer E, Belting HG, Affolter M, Lecaudey V, Mochizuki N. Flow-Dependent Endothelial YAP Regulation Contributes to Vessel Maintenance. Dev Cell 2017; 40:523-536.e6. [PMID: 28350986 DOI: 10.1016/j.devcel.2017.02.019] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/16/2017] [Accepted: 02/24/2017] [Indexed: 11/16/2022]
Abstract
Endothelial cells (ECs) line the inside of blood vessels and respond to mechanical cues generated by blood flow. Mechanical stimuli regulate the localization of YAP by reorganizing the actin cytoskeleton. Here we demonstrate blood-flow-mediated regulation of endothelial YAP in vivo. We indirectly monitored transcriptional activity of Yap1 (zebrafish YAP) and its spatiotemporal localization in living zebrafish and found that Yap1 entered the nucleus and promoted transcription in response to blood flow. In cultured human ECs, laminar shear stress induced nuclear import of YAP and its transcriptional activity in a manner independent of Hippo signaling. We uncovered a molecular mechanism by which flow induced the nuclear translocation of YAP through the regulation of filamentous actin and angiomotin. Yap1 mutant zebrafish showed a defect in vascular stability, indicating an essential role for Yap1 in blood vessels. Our data imply that endothelial Yap1 functions in response to flow to maintain blood vessels.
Collapse
Affiliation(s)
- Hiroyuki Nakajima
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Sobhika Agarwala
- Developmental Biology, SFB850, Institute for Biology I, Albert Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Kenta Terai
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Yoshidakonoe-cho, Sakyo-ku, Kyoto 606-8315, Japan
| | - Hajime Fukui
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Shigetomo Fukuhara
- Department of Molecular Pathophysiology, Nippon Medical School, Kawasaki, Kanagawa 211-8533, Japan
| | - Koji Ando
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Takahiro Miyazaki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Yasuhiro Yokota
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan
| | - Etienne Schmelzer
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Heinz-Georg Belting
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Markus Affolter
- Biozentrum der Universität Basel, Klingelbergstrasse 50/70, 4056 Basel, Switzerland
| | - Virginie Lecaudey
- Department of Developmental Biology of Vertebrates, Institute for Cell Biology and Neurosciences, Goethe University of Frankfurt, 60438 Frankfurt, Germany
| | - Naoki Mochizuki
- Department of Cell Biology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan; AMED-CREST, National Cerebral and Cardiovascular Center, 5-7-1, Suita, Osaka 565-8565, Japan.
| |
Collapse
|
199
|
Abstract
In the last 30 years, the zebrafish has become a widely used model organism for research on vertebrate development and disease. Through a powerful combination of genetics and experimental embryology, significant inroads have been made into the regulation of embryonic axis formation, organogenesis, and the development of neural networks. Research with this model has also expanded into other areas, including the genetic regulation of aging, regeneration, and animal behavior. Zebrafish are a popular model because of the ease with which they can be maintained, their small size and low cost, the ability to obtain hundreds of embryos on a daily basis, and the accessibility, translucency, and rapidity of early developmental stages. This primer describes the swift progress of genetic approaches in zebrafish and highlights recent advances that have led to new insights into vertebrate biology.
Collapse
|
200
|
Passoni G, Langevin C, Palha N, Mounce BC, Briolat V, Affaticati P, De Job E, Joly JS, Vignuzzi M, Saleh MC, Herbomel P, Boudinot P, Levraud JP. Imaging of viral neuroinvasion in the zebrafish reveals that Sindbis and chikungunya viruses favour different entry routes. Dis Model Mech 2017; 10:847-857. [PMID: 28483796 PMCID: PMC5536907 DOI: 10.1242/dmm.029231] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 05/02/2017] [Indexed: 12/13/2022] Open
Abstract
Alphaviruses, such as chikungunya virus (CHIKV) and Sindbis virus (SINV), are vector-borne pathogens that cause acute illnesses in humans and are sometimes associated with neuropathies, especially in infants and elderly patients. Little is known about their mechanism of entry into the central nervous system (CNS), even for SINV, which has been used extensively as a model for viral encephalopathies. We previously established a CHIKV infection model in the optically transparent zebrafish larva; here we describe a new SINV infection model in this host. We imaged in vivo the onset and progression of the infection caused by intravenous SINV inoculation. Similar to that described for CHIKV, infection in the periphery was detected early and was transient, whereas CNS infection started at later time points and was persistent or progressive. We then tested the possible mechanisms of neuroinvasion by CHIKV and SINV. Neither virus relied on macrophage-mediated transport to access the CNS. CHIKV, but not SINV, always infects endothelial cells of the brain vasculature. By contrast, axonal transport was much more efficient with SINV than CHIKV, both from the periphery to the CNS and between neural tissues. Thus, the preferred mechanisms of neuroinvasion by these two related viruses are distinct, providing a powerful imaging-friendly system to compare mechanisms and prevention methods of encephalopathies.
Collapse
Affiliation(s)
- Gabriella Passoni
- Virology and Molecular Immunology, INRA, Université Paris-Saclay, Domaine de Vilvert, Jouy-en-Josas F-78352, France.,Macrophages and Development of Immunity, Institut Pasteur, CNRS UMR 3738, 25 rue du docteur Roux, Paris F-75015, France
| | - Christelle Langevin
- Virology and Molecular Immunology, INRA, Université Paris-Saclay, Domaine de Vilvert, Jouy-en-Josas F-78352, France
| | - Nuno Palha
- Macrophages and Development of Immunity, Institut Pasteur, CNRS UMR 3738, 25 rue du docteur Roux, Paris F-75015, France
| | - Bryan C Mounce
- Viral Populations and Pathogenesis Unit, Institut Pasteur, CNRS UMR 3569, Paris F-75015, France
| | - Valérie Briolat
- Macrophages and Development of Immunity, Institut Pasteur, CNRS UMR 3738, 25 rue du docteur Roux, Paris F-75015, France
| | - Pierre Affaticati
- Tefor Core Facility, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris-Saclay, Gif-sur-Yvette F-91190, France
| | - Elodie De Job
- Tefor Core Facility, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris-Saclay, Gif-sur-Yvette F-91190, France
| | - Jean-Stéphane Joly
- Tefor Core Facility, Paris-Saclay Institute of Neuroscience, CNRS, Université Paris-Saclay, Gif-sur-Yvette F-91190, France
| | - Marco Vignuzzi
- Viral Populations and Pathogenesis Unit, Institut Pasteur, CNRS UMR 3569, Paris F-75015, France
| | - Maria-Carla Saleh
- Viruses and RNA Interference, Institut Pasteur, CNRS UMR 3569, Paris F-75015, France
| | - Philippe Herbomel
- Macrophages and Development of Immunity, Institut Pasteur, CNRS UMR 3738, 25 rue du docteur Roux, Paris F-75015, France
| | - Pierre Boudinot
- Virology and Molecular Immunology, INRA, Université Paris-Saclay, Domaine de Vilvert, Jouy-en-Josas F-78352, France
| | - Jean-Pierre Levraud
- Macrophages and Development of Immunity, Institut Pasteur, CNRS UMR 3738, 25 rue du docteur Roux, Paris F-75015, France
| |
Collapse
|