1
|
Li Y, Li Y, Huang B, Zhang R, He J, Luo L, Yang Y. Long-term labelling and tracing of endodermal cells using a perpetual cycling Gal4-UAS system. Development 2025; 152:dev204289. [PMID: 40116142 PMCID: PMC11959616 DOI: 10.1242/dev.204289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 02/12/2025] [Indexed: 03/22/2025]
Abstract
Cell labelling and lineage tracing are indispensable tools in developmental biology, offering powerful means with which to visualise and understand the complex dynamics of cell populations during embryogenesis. Traditional cell labelling relies heavily on signal stability, promoter strength and stage specificity, limiting its application in long-term tracing. In this report, we optimise and reconfigure a perpetual cycling Gal4-UAS system employing a previously unreported Gal4 fusion protein and the autoregulatory Gal4 expression loop. As validated through heat-shock induction, this configuration ensures sustained transcription of reporter genes in target cells and their descendant cells while minimising cytotoxicity, thereby achieving long-term labelling and tracing. Further exploiting this system, we generate zebrafish transgenic lines with continuous fluorescent labelling specific to the endoderm, and demonstrate its effectiveness in long-term tracing by showing the progression of endoderm development from embryo to adult, providing visualisation of endodermal cells and their derived tissues. This continuous labelling and tracing strategy can span the entire process of endodermal differentiation, from progenitor cells to mature functional cells, and is applicable to studying endoderm patterning and organogenesis.
Collapse
Affiliation(s)
- Yanfeng Li
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - You Li
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Bangzhuo Huang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Ruhao Zhang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Jianbo He
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
- School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yun Yang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei 400715, Chongqing, China
| |
Collapse
|
2
|
de Calbiac H, Renault S, Haouy G, Jung V, Roger K, Zhou Q, Campanari ML, Chentout L, Demy DL, Marian A, Goudin N, Edbauer D, Guerrera C, Ciura S, Kabashi E. Poly-GP accumulation due to C9orf72 loss of function induces motor neuron apoptosis through autophagy and mitophagy defects. Autophagy 2024; 20:2164-2185. [PMID: 39316747 PMCID: PMC11423671 DOI: 10.1080/15548627.2024.2358736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 09/26/2024] Open
Abstract
The GGGGCC hexanucleotide repeat expansion (HRE) of the C9orf72 gene is the most frequent cause of amyotrophic lateral sclerosis (ALS), a devastative neurodegenerative disease characterized by motor neuron degeneration. C9orf72 HRE is associated with lowered levels of C9orf72 expression and its translation results in the production of dipeptide-repeats (DPRs). To recapitulate C9orf72-related ALS disease in vivo, we developed a zebrafish model where we expressed glycine-proline (GP) DPR in a c9orf72 knockdown context. We report that C9orf72 gain- and loss-of-function properties act synergistically to induce motor neuron degeneration and paralysis with poly(GP) accumulating preferentially within motor neurons along with Sqstm1/p62 aggregation indicating macroautophagy/autophagy deficits. Poly(GP) levels were shown to accumulate upon c9orf72 downregulation and were comparable to levels assessed in autopsy samples of patients carrying C9orf72 HRE. Chemical boosting of autophagy using rapamycin or apilimod, is able to rescue motor deficits. Proteomics analysis of zebrafish-purified motor neurons unravels mitochondria dysfunction confirmed through a comparative analysis of previously published C9orf72 iPSC-derived motor neurons. Consistently, 3D-reconstructions of motor neuron demonstrate that poly(GP) aggregates colocalize to mitochondria, thus inducing their elongation and swelling and the failure of their processing by mitophagy, with mitophagy activation through urolithin A preventing locomotor deficits. Finally, we report apoptotic-related increased amounts of cleaved Casp3 (caspase 3, apoptosis-related cysteine peptidase) and rescue of motor neuron degeneration by constitutive inhibition of Casp9 or treatment with decylubiquinone. Here we provide evidence of key pathogenic steps in C9ALS-FTD that can be targeted through pharmacological avenues, thus raising new therapeutic perspectives for ALS patients.
Collapse
Affiliation(s)
- Hortense de Calbiac
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Solène Renault
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Grégoire Haouy
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Vincent Jung
- Proteomics Platform 3P5Necker, INSERM US24/CNRS UMS, Paris Descartes University, Structure Fédérative de Recherche Necker, Paris, France
| | - Kevin Roger
- Proteomics Platform 3P5Necker, INSERM US24/CNRS UMS, Paris Descartes University, Structure Fédérative de Recherche Necker, Paris, France
| | - Qihui Zhou
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Maria-Letizia Campanari
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Loïc Chentout
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Doris Lou Demy
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Anca Marian
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Nicolas Goudin
- Imaging Core Facility, INSERM US24/CNRS UMS3633, Paris, France
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
- Ludwig-Maximilians-Universität (LMU) Munich, Graduate School of Systemic Neurosciences (GSN), Munich, Germany
| | - Chiara Guerrera
- Proteomics Platform 3P5Necker, INSERM US24/CNRS UMS, Paris Descartes University, Structure Fédérative de Recherche Necker, Paris, France
| | - Sorana Ciura
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| | - Edor Kabashi
- Imagine Institute, INSERM UMR 1163, Team Translational Research for Neurological Diseases, Paris Descartes University, Paris, France
| |
Collapse
|
3
|
Chambers BE, Weaver NE, Lara CM, Nguyen TK, Wingert RA. (Zebra)fishing for nephrogenesis genes. Tissue Barriers 2024; 12:2219605. [PMID: 37254823 PMCID: PMC11042071 DOI: 10.1080/21688370.2023.2219605] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/14/2023] [Indexed: 06/01/2023] Open
Abstract
Kidney disease is a devastating condition affecting millions of people worldwide, where over 100,000 patients in the United States alone remain waiting for a lifesaving organ transplant. Concomitant with a surge in personalized medicine, single-gene mutations, and polygenic risk alleles have been brought to the forefront as core causes of a spectrum of renal disorders. With the increasing prevalence of kidney disease, it is imperative to make substantial strides in the field of kidney genetics. Nephrons, the core functional units of the kidney, are epithelial tubules that act as gatekeepers of body homeostasis by absorbing and secreting ions, water, and small molecules to filter the blood. Each nephron contains a series of proximal and distal segments with explicit metabolic functions. The embryonic zebrafish provides an ideal platform to systematically dissect the genetic cues governing kidney development. Here, we review the use of zebrafish to discover nephrogenesis genes.
Collapse
Affiliation(s)
- Brooke E. Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Nicole E. Weaver
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Caroline M. Lara
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| |
Collapse
|
4
|
Rahman MR, Kawabe Y, Suzuki K, Chen S, Amamoto Y, Kamihira M. Inducible transgene expression in CHO cells using an artificial transcriptional activator with estrogen-binding domain. Biotechnol J 2024; 19:e2300362. [PMID: 38161242 DOI: 10.1002/biot.202300362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 12/04/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Biopharmaceuticals, including therapeutic antibodies, are rapidly growing products in the pharmaceutical market. Mammalian cells, such as Chinese hamster ovary (CHO) cells, are widely used as production hosts because recombinant antibodies require complex three-dimensional structures modified with sugar chains. Recombinant protein production using mammalian cells is generally performed with cell growth. In this study, we developed a technology that controls cell growth and recombinant protein production to induce recombinant protein production with predetermined timing. Expression of green fluorescent protein (GFP) gene and a single-chain antibody fused with the Fc-region of the human IgG1 (scFv-Fc) gene can be induced and mediated by the estrogen receptor-based artificial transcription factor Gal4-ERT2-VP16 and corresponding inducer drugs. We generated CHO cells using an artificial gene expression system. The addition of various concentrations of inducer drugs to the culture medium allowed control of proliferation and transgene expression of the engineered CHO cells. Use of 4-hydroxytamoxifen, an antagonist of estrogen, as an inducing agent yielded high gene expression at a concentration more than 10-fold lower than that of β-estradiol. When scFv-Fc was produced under inducing conditions, continuous production was possible for more than 2 weeks while maintaining high specific productivity (57 pg cell-1 day-1 ). This artificial gene expression control system that utilizes the estrogen response of estrogen receptors can be an effective method for inducible production of biopharmaceuticals.
Collapse
Affiliation(s)
- Md Rashidur Rahman
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Kozumi Suzuki
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Satoshi Chen
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Yuki Amamoto
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| |
Collapse
|
5
|
Zhu R, Santat LA, Markson JS, Nandagopal N, Gregrowicz J, Elowitz MB. Reconstitution of morphogen shuttling circuits. SCIENCE ADVANCES 2023; 9:eadf9336. [PMID: 37436981 PMCID: PMC10337948 DOI: 10.1126/sciadv.adf9336] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 06/08/2023] [Indexed: 07/14/2023]
Abstract
Developing tissues form spatial patterns by establishing concentration gradients of diffusible signaling proteins called morphogens. The bone morphogenetic protein (BMP) morphogen pathway uses a family of extracellular modulators to reshape signaling gradients by actively "shuttling" ligands to different locations. It has remained unclear what circuits are sufficient to enable shuttling, what other patterns they can generate, and whether shuttling is evolutionarily conserved. Here, using a synthetic, bottom-up approach, we compared the spatiotemporal dynamics of different extracellular circuits. Three proteins-Chordin, Twsg, and the BMP-1 protease-successfully displaced gradients by shuttling ligands away from the site of production. A mathematical model explained the different spatial dynamics of this and other circuits. Last, combining mammalian and Drosophila components in the same system suggests that shuttling is a conserved capability. Together, these results reveal principles through which extracellular circuits control the spatiotemporal dynamics of morphogen signaling.
Collapse
Affiliation(s)
- Ronghui Zhu
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Leah A. Santat
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Joseph S. Markson
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | - Jan Gregrowicz
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Michael B. Elowitz
- Howard Hughes Medical Institute and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
6
|
Schlotawa L, Lopez A, Sanchez-Elexpuru G, Tyrkalska SD, Rubinsztein DC, Fleming A. An inducible expression system for the manipulation of autophagic flux in vivo. Autophagy 2023; 19:1582-1595. [PMID: 36310368 PMCID: PMC10240996 DOI: 10.1080/15548627.2022.2135824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 01/18/2023] Open
Abstract
Much of our understanding of the intracellular regulation of macroautophagy/autophagy comes from in vitro studies. However, there remains a paucity of knowledge about how this process is regulated within different tissues during development, aging and disease in vivo. Because upregulation of autophagy is considered a promising therapeutic strategy for the treatment of diverse disorders, it is vital that we understand how this pathway functions in different tissues and this is best done by in vivo analysis. Similarly, to understand the role of autophagy in the pathogenesis of disease, it is important to study this process in the whole animal to investigate how tissue-specific changes in flux and cell-autonomous versus non-cell-autonomous effects alter disease progression. To this end, we have developed an inducible expression system to up- or downregulate autophagy in vivo, in zebrafish. We have used a modified version of the Gal4-UAS expression system to allow inducible expression of autophagy up- or downregulating transgenes by addition of tamoxifen. Using this inducible expression system, we have tested which transgenes robustly up- or downregulate autophagy and have validated these tools using Lc3-II blots and puncta analysis and disease rescue in a zebrafish model of neurodegeneration. These tools allow the temporal control of autophagy via the administration of tamoxifen and spatial control via tissue or cell-specific ERT2-Gal4 driver lines and will enable the investigation of how cell- or tissue-specific changes in autophagic flux affect processes such as aging, inflammation and neurodegeneration in vivo.Abbreviations: ANOVA: analysis of variance; Atg: autophagy related; Bcl2l11/Bim: BCL2 like 11; d.p.f.: days post-fertilization; Cryaa: crystallin, alpha a: DMSO: dimethyl sulfoxide; Elavl3: ELAV like neuron-specific RNA binding protein 3; ER: estrogen receptor; ERT2: modified ligand-binding domain of human ESR1/estrogen receptor α; Gal4: galactose-responsive transcription factor 4; GFP: green fluorescent protein; h.p.f.: hours post-fertilization; HSP: heat-shock protein; Map1lc3/Lc3: microtubule-associated protein 1 light chain 3; RFP: red fluorescent protein; SD: standard deviation; SEM: standard error of the mean; UAS: upstream activating sequence; Ubb: ubiquitin b.
Collapse
Affiliation(s)
- Lars Schlotawa
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical, Research, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Ana Lopez
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical, Research, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Gentzane Sanchez-Elexpuru
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical, Research, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sylwia D. Tyrkalska
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical, Research, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - David C. Rubinsztein
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical, Research, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, the Keith Peters Building, Cambridge, UK
| | - Angeleen Fleming
- Department of Medical Genetics, University of Cambridge, Cambridge Institute for Medical, Research, Cambridge, UK
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge Institute for Medical Research, the Keith Peters Building, Cambridge, UK
| |
Collapse
|
7
|
Nikolaou N, Gordon PM, Hamid F, Taylor R, Lloyd-Jones J, Makeyev EV, Houart C. Cytoplasmic pool of U1 spliceosome protein SNRNP70 shapes the axonal transcriptome and regulates motor connectivity. Curr Biol 2022; 32:5099-5115.e8. [PMID: 36384140 DOI: 10.1016/j.cub.2022.10.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 09/09/2022] [Accepted: 10/21/2022] [Indexed: 11/17/2022]
Abstract
Regulation of pre-mRNA splicing and polyadenylation plays a profound role in neurons by diversifying the proteome and modulating gene expression in response to physiological cues. Although most of the pre-mRNA processing is thought to occur in the nucleus, numerous splicing regulators are also found in neurites. Here, we show that U1-70K/SNRNP70, a component of the major spliceosome, localizes in RNA-associated granules in zebrafish axons. We identify the extra-nuclear SNRNP70 as an important regulator of motor axonal growth, nerve-dependent acetylcholine receptor (AChR) clustering, and neuromuscular synaptogenesis. This cytoplasmic pool has a protective role for a limited number of transcripts regulating their abundance and trafficking inside axons. Moreover, non-nuclear SNRNP70 regulates splice variants of transcripts such as agrin, thereby controlling synapse formation. Our results point to an unexpected, yet essential, function of non-nuclear SNRNP70 in axonal development, indicating a role of spliceosome proteins in cytoplasmic RNA metabolism during neuronal connectivity.
Collapse
Affiliation(s)
- Nikolas Nikolaou
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK; Department of Life Sciences, University of Bath, Bath BA2 7AY, UK.
| | - Patricia M Gordon
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Richard Taylor
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK
| | | | - Eugene V Makeyev
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Corinne Houart
- Centre for Developmental Neurobiology MRC CNDD, IoPPN, Guy's Campus, King's College London, London SE1 1UL, UK.
| |
Collapse
|
8
|
Labbaf Z, Petratou K, Ermlich L, Backer W, Tarbashevich K, Reichman-Fried M, Luschnig S, Schulte-Merker S, Raz E. A robust and tunable system for targeted cell ablation in developing embryos. Dev Cell 2022; 57:2026-2040.e5. [PMID: 35914525 DOI: 10.1016/j.devcel.2022.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 05/10/2022] [Accepted: 07/07/2022] [Indexed: 11/03/2022]
Abstract
Cell ablation is a key method in the research fields of developmental biology, tissue regeneration, and tissue homeostasis. Eliminating specific cell populations allows for characterizing interactions that control cell differentiation, death, behavior, and spatial organization of cells. Current methodologies for inducing cell death suffer from relatively slow kinetics, making them unsuitable for analyzing rapid events and following primary and immediate consequences of the ablation. To address this, we developed a cell-ablation system that is based on bacterial toxin/anti-toxin proteins and enables rapid and cell-autonomous elimination of specific cell types and organs in zebrafish embryos. A unique feature of this system is that it uses an anti-toxin, which allows for controlling the degree and timing of ablation and the resulting phenotypes. The transgenic zebrafish generated in this work represent a highly efficient tool for cell ablation, and this approach is applicable to other model organisms as demonstrated here for Drosophila.
Collapse
Affiliation(s)
- Zahra Labbaf
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster 48149, Germany
| | - Kleio Petratou
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Münster 48149, Germany
| | - Laura Ermlich
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster 48149, Germany
| | - Wilko Backer
- Institute for Integrative Cell Biology and Physiology, University of Münster, Münster 48149, Germany
| | - Katsiaryna Tarbashevich
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster 48149, Germany
| | - Michal Reichman-Fried
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster 48149, Germany
| | - Stefan Luschnig
- Institute for Integrative Cell Biology and Physiology, University of Münster, Münster 48149, Germany
| | - Stefan Schulte-Merker
- Institute for Cardiovascular Organogenesis and Regeneration, Faculty of Medicine, Münster 48149, Germany
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster 48149, Germany.
| |
Collapse
|
9
|
A New Zebrafish Model to Measure Neuronal α-Synuclein Clearance In Vivo. Genes (Basel) 2022; 13:genes13050868. [PMID: 35627253 PMCID: PMC9141618 DOI: 10.3390/genes13050868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023] Open
Abstract
The accumulation and aggregation of α-synuclein (α-SYN) is a common characteristic of synucleinopathies, such as Parkinson's Disease (PD), Dementia with Lewy Bodies (DLB) or Multiple System Atrophy (MSA). Multiplications of the wildtype gene of α-SYN (SNCA) and most point mutations make α-SYN more aggregate-prone, and are associated with mitochondrial defects, trafficking obstruction, and impaired proteostasis, which contribute to elevated neuronal death. Here, we present new zebrafish models expressing either human wildtype (wt), or A53T mutant, α-SYN that recapitulate the above-mentioned hallmarks of synucleinopathies. The appropriate clearance of toxic α-SYN has been previously shown to play a key role in maintaining cell homeostasis and survival. However, the paucity of models to investigate α-SYN degradation in vivo limits our understanding of this process. Based on our recently described imaging method for measuring tau protein clearance in neurons in living zebrafish, we fused human SNCA to the photoconvertible protein Dendra2 which enabled analyses of wt and A53T α-SYN clearance kinetics in vivo. Moreover, these zebrafish models can be used to investigate the kinetics of α-SYN aggregation and to study the mechanisms, and potential new targets, controlling the clearance of both soluble and aggregated α-SYN.
Collapse
|
10
|
Zhu R, Del Rio-Salgado JM, Garcia-Ojalvo J, Elowitz MB. Synthetic multistability in mammalian cells. Science 2022; 375:eabg9765. [PMID: 35050677 DOI: 10.1126/science.abg9765] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In multicellular organisms, gene regulatory circuits generate thousands of molecularly distinct, mitotically heritable states through the property of multistability. Designing synthetic multistable circuits would provide insight into natural cell fate control circuit architectures and would allow engineering of multicellular programs that require interactions among distinct cell types. We created MultiFate, a naturally inspired, synthetic circuit that supports long-term, controllable, and expandable multistability in mammalian cells. MultiFate uses engineered zinc finger transcription factors that transcriptionally self-activate as homodimers and mutually inhibit one another through heterodimerization. Using a model-based design, we engineered MultiFate circuits that generate as many as seven states, each stable for at least 18 days. MultiFate permits controlled state switching and modulation of state stability through external inputs and can be expanded with additional transcription factors. These results provide a foundation for engineering multicellular behaviors in mammalian cells.
Collapse
Affiliation(s)
- Ronghui Zhu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jesus M Del Rio-Salgado
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jordi Garcia-Ojalvo
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.,Howard Hughes Medical Institute, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
11
|
Gorelick DA, Lucia C, Hao R, Karim S, Bondesson M. Use of Reporter Genes to Analyze Estrogen Response: The Transgenic Zebrafish Model. Methods Mol Biol 2022; 2418:173-185. [PMID: 35119666 DOI: 10.1007/978-1-0716-1920-9_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In vivo models to detect estrogenic compounds are very valuable for screening for endocrine disruptors. Here we describe the use of transgenic estrogen reporter zebrafish as an in vivo model for the identification of estrogenic properties of compounds. Live imaging of these transgenic fish provides knowledge of estrogen receptor specificity of different ligands as well as dynamics of estrogen signaling. Coupled to image analysis, the model can provide quantitative concentration-response information on estrogenic activity of chemical compounds.
Collapse
Affiliation(s)
- Daniel A Gorelick
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Caroline Lucia
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
| | - Ruixin Hao
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA
| | - Silvia Karim
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA.
| |
Collapse
|
12
|
IQ-Switch is a QF-based innocuous, silencing-free, and inducible gene switch system in zebrafish. Commun Biol 2021; 4:1405. [PMID: 34916605 PMCID: PMC8677817 DOI: 10.1038/s42003-021-02923-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/24/2021] [Indexed: 11/08/2022] Open
Abstract
Though various transgene expression switches have been adopted in a wide variety of organisms for basic and biomedical research, intrinsic obstacles of those existing systems, including toxicity and silencing, have been limiting their use in vertebrate transgenesis. Here we demonstrate a novel QF-based binary transgene switch (IQ-Switch) that is relatively free of driver toxicity and transgene silencing, and exhibits potent and highly tunable transgene activation by the chemical inducer tebufenozide, a non-toxic lipophilic molecule to developing zebrafish with negligible background. The interchangeable IQ-Switch makes it possible to elicit ubiquitous and tissue specific transgene expression in a spatiotemporal manner. We generated a RASopathy disease model using IQ-Switch and demonstrated that the RASopathy symptoms were ameliorated by the specific BRAF(V600E) inhibitor vemurafenib, validating the therapeutic use of the gene switch. The orthogonal IQ-Switch provides a state-of-the-art platform for flexible regulation of transgene expression in zebrafish, potentially applicable in cell-based systems and other model organisms.
Collapse
|
13
|
Reconstitution of Morphogen Signaling Gradients in Cultured Cells. Methods Mol Biol 2020. [PMID: 33340353 DOI: 10.1007/978-1-0716-1174-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Development of multicellular organisms depends on the proper establishment of signaling information in space and time. Secreted molecules called morphogens form concentration gradients in space and provide positional information to differentiating cells within the organism. Although the key molecular components of morphogen pathways have been identified, how the architectures and key parameters of morphogen pathways control the properties of signaling gradients, such as their size, speed, and robustness to perturbations, remains challenging to study in developing embryos. Reconstituting morphogen gradients in cell culture provides an alternative approach to address this question. Here we describe the methodology for reconstituting Sonic Hedgehog (SHH) signaling gradients in mouse fibroblast cells. The protocol includes the design of morphogen sending and receiving cell lines, the setup of radial and linear gradients, the quantitative time-lapse imaging, and the data analysis. Similar approaches could potentially be applied to other cell-cell communication pathways.
Collapse
|
14
|
Dalle Nogare DE, Natesh N, Vishwasrao HD, Shroff H, Chitnis AB. Zebrafish Posterior Lateral Line primordium migration requires interactions between a superficial sheath of motile cells and the skin. eLife 2020; 9:58251. [PMID: 33237853 PMCID: PMC7688310 DOI: 10.7554/elife.58251] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
The Zebrafish Posterior Lateral Line primordium migrates in a channel between the skin and somites. Its migration depends on the coordinated movement of its mesenchymal-like leading cells and trailing cells, which form epithelial rosettes, or protoneuromasts. We describe a superficial population of flat primordium cells that wrap around deeper epithelialized cells and extend polarized lamellipodia to migrate apposed to the overlying skin. Polarization of lamellipodia extended by both superficial and deeper protoneuromast-forming cells depends on Fgf signaling. Removal of the overlying skin has similar effects on superficial and deep cells: lamellipodia are lost, blebs appear instead, and collective migration fails. When skinned embryos are embedded in Matrigel, basal and superficial lamellipodia are recovered; however, only the directionality of basal protrusions is recovered, and migration is not rescued. These observations support a key role played by superficial primordium cells and the skin in directed migration of the Posterior Lateral Line primordium.
Collapse
Affiliation(s)
- Damian E Dalle Nogare
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Naveen Natesh
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Harshad D Vishwasrao
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, United States
| | - Hari Shroff
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, United States.,Laboratory of High Resolution Optical Imaging, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, United States
| | - Ajay B Chitnis
- Section on Neural Developmental Dynamics, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| |
Collapse
|
15
|
Freudenblum J, Meyer D, Kimmel RA. Inducible Mosaic Cell Labeling Provides Insights Into Pancreatic Islet Morphogenesis. Front Cell Dev Biol 2020; 8:586651. [PMID: 33102488 PMCID: PMC7546031 DOI: 10.3389/fcell.2020.586651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/02/2020] [Indexed: 11/13/2022] Open
Abstract
Pancreatic islets, discrete microorgans embedded within the exocrine pancreas, contain beta cells which are critical for glucose homeostasis. Loss or dysfunction of beta cells leads to diabetes, a disease with expanding global prevalence, and for which regenerative therapies are actively being pursued. Recent efforts have focused on producing mature beta cells in vitro, but it is increasingly recognized that achieving a faithful three-dimensional islet structure is crucial for generating fully functional beta cells. Our current understanding of islet morphogenesis is far from complete, due to the deep internal location of the pancreas in mammalian models, which hampers direct visualization. Zebrafish is a model system well suited for studies of pancreas morphogenesis due to its transparency and the accessible location of the larval pancreas. In order to further clarify the cellular mechanisms of islet formation, we have developed new tools for in vivo visualization of single-cell dynamics. Our results show that clustering islet cells make contact and interconnect through dynamic actin-rich processes, move together while remaining in close proximity to the duct, and maintain high protrusive motility after forming clusters. Quantitative analyses of cell morphology and motility in 3-dimensions lays the groundwork to define therapeutically applicable factors responsible for orchestrating the morphogenic behaviors of coalescing endocrine cells.
Collapse
Affiliation(s)
- Julia Freudenblum
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Dirk Meyer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology/CMBI, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
16
|
McCallum S, Obata Y, Fourli E, Boeing S, Peddie CJ, Xu Q, Horswell S, Kelsh RN, Collinson L, Wilkinson D, Pin C, Pachnis V, Heanue TA. Enteric glia as a source of neural progenitors in adult zebrafish. eLife 2020; 9:e56086. [PMID: 32851974 PMCID: PMC7521928 DOI: 10.7554/elife.56086] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 08/26/2020] [Indexed: 12/23/2022] Open
Abstract
The presence and identity of neural progenitors in the enteric nervous system (ENS) of vertebrates is a matter of intense debate. Here, we demonstrate that the non-neuronal ENS cell compartment of teleosts shares molecular and morphological characteristics with mammalian enteric glia but cannot be identified by the expression of canonical glial markers. However, unlike their mammalian counterparts, which are generally quiescent and do not undergo neuronal differentiation during homeostasis, we show that a relatively high proportion of zebrafish enteric glia proliferate under physiological conditions giving rise to progeny that differentiate into enteric neurons. We also provide evidence that, similar to brain neural stem cells, the activation and neuronal differentiation of enteric glia are regulated by Notch signalling. Our experiments reveal remarkable similarities between enteric glia and brain neural stem cells in teleosts and open new possibilities for use of mammalian enteric glia as a potential source of neurons to restore the activity of intestinal neural circuits compromised by injury or disease.
Collapse
Affiliation(s)
- Sarah McCallum
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Yuuki Obata
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Evangelia Fourli
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Stefan Boeing
- Bionformatics & Biostatistics Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Christopher J Peddie
- Electron Microscopy Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Qiling Xu
- Neural Development Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Stuart Horswell
- Bionformatics & Biostatistics Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - Robert N Kelsh
- Department of Biology and Biochemistry, University of BathBathUnited Kingdom
| | - Lucy Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick InstituteLondonUnited Kingdom
| | - David Wilkinson
- Neural Development Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Carmen Pin
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology and Safety Sciences, R&D, AstraZenecaCambridgeUnited Kingdom
| | - Vassilis Pachnis
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| | - Tiffany A Heanue
- Development and Homeostasis of the Nervous System Laboratory, The Francis Crick InstituteLondonUnited Kingdom
| |
Collapse
|
17
|
Rosowski EE. Determining macrophage versus neutrophil contributions to innate immunity using larval zebrafish. Dis Model Mech 2020; 13:13/1/dmm041889. [PMID: 31932292 PMCID: PMC6994940 DOI: 10.1242/dmm.041889] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The specific roles of the two major innate immune cell types – neutrophils and macrophages – in response to infection and sterile inflammation are areas of great interest. The larval zebrafish model of innate immunity, and the imaging capabilities it provides, is a source of new research and discoveries in this field. Multiple methods have been developed in larval zebrafish to specifically deplete functional macrophages or neutrophils. Each of these has pros and cons, as well as caveats, that often make it difficult to directly compare results from different studies. The purpose of this Review is to (1) explore the pros, cons and caveats of each of these immune cell-depleted models; (2) highlight and place into a broader context recent key findings on the specific functions of innate immune cells using these models; and (3) explore future directions in which immune cell depletion methods are being expanded. Summary: Macrophages and neutrophils are distinct innate immune cells with diverse roles in diverse inflammatory contexts. Recent research in larval zebrafish using cell-specific depletion methods has revealed new insights into these cells' functions.
Collapse
Affiliation(s)
- Emily E Rosowski
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
18
|
Vilches-Moure JG. Embryonic Chicken ( Gallus gallus domesticus) as a Model of Cardiac Biology and Development. Comp Med 2019; 69:184-203. [PMID: 31182184 PMCID: PMC6591676 DOI: 10.30802/aalas-cm-18-000061] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/06/2018] [Accepted: 11/29/2018] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease remains one of the top contributors to morbidity and mortality in the United States. Increasing evidence suggests that many processes, pathways, and programs observed during development and organogenesis are recapitulated in adults in the face of disease. Therefore, a heightened understanding of cardiac development and organogenesis will help increase our understanding of developmental defects and cardiovascular diseases in adults. Chicks have long served as a model system in which to study developmental problems. Detailed descriptions of morphogenesis, low cost, accessibility, ease of manipulation, and the optimization of genetic engineering techniques have made chicks a robust model for studying development and make it a powerful platform for cardiovascular research. This review summarizes the cardiac developmental milestones of embryonic chickens, practical considerations when working with chicken embryos, and techniques available for use in chicks (including tissue chimeras, genetic manipulations, and live imaging). In addition, this article highlights examples that accentuate the utility of the embryonic chicken as model system in which to study cardiac development, particularly epicardial development, and that underscore the importance of how studying development informs our understanding of disease.
Collapse
Affiliation(s)
- José G Vilches-Moure
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, California,
| |
Collapse
|
19
|
Gu Q, Yang X, Lv J, Zhang J, Xia B, Kim JD, Wang R, Xiong F, Meng S, Clements TP, Tandon B, Wagner DS, Diaz MF, Wenzel PL, Miller YI, Traver D, Cooke JP, Li W, Zon LI, Chen K, Bai Y, Fang L. AIBP-mediated cholesterol efflux instructs hematopoietic stem and progenitor cell fate. Science 2019; 363:1085-1088. [PMID: 30705153 DOI: 10.1126/science.aav1749] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 01/22/2019] [Indexed: 12/18/2022]
Abstract
Hypercholesterolemia, the driving force of atherosclerosis, accelerates the expansion and mobilization of hematopoietic stem and progenitor cells (HSPCs). The molecular determinants connecting hypercholesterolemia with hematopoiesis are unclear. Here, we report that a somite-derived prohematopoietic cue, AIBP, orchestrates HSPC emergence from the hemogenic endothelium, a type of specialized endothelium manifesting hematopoietic potential. Mechanistically, AIBP-mediated cholesterol efflux activates endothelial Srebp2, the master transcription factor for cholesterol biosynthesis, which in turn transactivates Notch and promotes HSPC emergence. Srebp2 inhibition impairs hypercholesterolemia-induced HSPC expansion. Srebp2 activation and Notch up-regulation are associated with HSPC expansion in hypercholesterolemic human subjects. Genome-wide chromatin immunoprecipitation followed by sequencing (ChIP-seq), RNA sequencing (RNA-seq), and assay for transposase-accessible chromatin using sequencing (ATAC-seq) indicate that Srebp2 transregulates Notch pathway genes required for hematopoiesis. Our studies outline an AIBP-regulated Srebp2-dependent paradigm for HSPC emergence in development and HPSC expansion in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Qilin Gu
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA
| | - Xiaojie Yang
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA
| | - Jie Lv
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA.,Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA
| | - Jiaxiong Zhang
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA.,Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Bo Xia
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA.,Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA
| | - Jun-Dae Kim
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA
| | - Ruoyu Wang
- Department of Biochemistry and Molecular Biology, UTHealth McGovern Medical School, University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA.,Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Feng Xiong
- Department of Biochemistry and Molecular Biology, UTHealth McGovern Medical School, University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Shu Meng
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA
| | | | - Bhavna Tandon
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| | - Daniel S Wagner
- Department of BioSciences, Rice University, Houston, TX 77005, USA
| | - Miguel F Diaz
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Pamela L Wenzel
- Children's Regenerative Medicine Program, Department of Pediatric Surgery, Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yury I Miller
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - David Traver
- Division of Biological Sciences, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - John P Cooke
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA.,Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA.,Department of Cardiothoracic Surgeries, Weill Cornell Medical College, Cornell University, Ithaca, NY 10065, USA
| | - Wenbo Li
- Department of Biochemistry and Molecular Biology, UTHealth McGovern Medical School, University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA.,Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth Houston, Houston, TX 77030, USA
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kaifu Chen
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA. .,Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA.,Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA.,Department of Cardiothoracic Surgeries, Weill Cornell Medical College, Cornell University, Ithaca, NY 10065, USA
| | - Yongping Bai
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China.
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA. .,Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA.,Department of Cardiothoracic Surgeries, Weill Cornell Medical College, Cornell University, Ithaca, NY 10065, USA.,Department of Obstetrics and Gynecology, Houston Methodist, 6550 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
20
|
Li P, Markson JS, Wang S, Chen S, Vachharajani V, Elowitz MB. Morphogen gradient reconstitution reveals Hedgehog pathway design principles. Science 2018; 360:543-548. [PMID: 29622726 DOI: 10.1126/science.aao0645] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 03/22/2018] [Indexed: 12/30/2022]
Abstract
In developing tissues, cells estimate their spatial position by sensing graded concentrations of diffusible signaling proteins called morphogens. Morphogen-sensing pathways exhibit diverse molecular architectures, whose roles in controlling patterning dynamics and precision have been unclear. In this work, combining cell-based in vitro gradient reconstitution, genetic rewiring, and mathematical modeling, we systematically analyzed the distinctive architectural features of the Sonic Hedgehog pathway. We found that the combination of double-negative regulatory logic and negative feedback through the PTCH receptor accelerates gradient formation and improves robustness to variation in the morphogen production rate compared with alternative designs. The ability to isolate morphogen patterning from concurrent developmental processes and to compare the patterning behaviors of alternative, rewired pathway architectures offers a powerful way to understand and engineer multicellular patterning.
Collapse
Affiliation(s)
- Pulin Li
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Joseph S Markson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sheng Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Siheng Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Vipul Vachharajani
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michael B Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA. .,Howard Hughes Medical Institute and Department of Applied Physics, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
21
|
Schenk H, Müller-Deile J, Kinast M, Schiffer M. Disease modeling in genetic kidney diseases: zebrafish. Cell Tissue Res 2017; 369:127-141. [PMID: 28331970 DOI: 10.1007/s00441-017-2593-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/22/2017] [Indexed: 01/07/2023]
Abstract
Growing numbers of translational genomics studies are based on the highly efficient and versatile zebrafish (Danio rerio) vertebrate model. The increasing types of zebrafish models have improved our understanding of inherited kidney diseases, since they not only display pathophysiological changes but also give us the opportunity to develop and test novel treatment options in a high-throughput manner. New paradigms in inherited kidney diseases have been developed on the basis of the distinct genome conservation of approximately 70 % between zebrafish and humans in terms of existing gene orthologs. Several options are available to determine the functional role of a specific gene or gene sets. Permanent genome editing can be induced via complete gene knockout by using the CRISPR/Cas-system, among others, or via transient modification by using various morpholino techniques. Cross-species rescues succeeding knockdown techniques are employed to determine the functional significance of a target gene or a specific mutation. This article summarizes the current techniques and discusses their perspectives.
Collapse
Affiliation(s)
- Heiko Schenk
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Janina Müller-Deile
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Mark Kinast
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Mario Schiffer
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany.
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA.
| |
Collapse
|
22
|
Gorelick DA, Pinto CL, Hao R, Bondesson M. Use of Reporter Genes to Analyze Estrogen Response: The Transgenic Zebrafish Model. Methods Mol Biol 2016; 1366:315-325. [PMID: 26585145 DOI: 10.1007/978-1-4939-3127-9_24] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
In vivo models to detect estrogenic compounds are very valuable for screening for endocrine disruptors. Here we describe the use of transgenic estrogen reporter zebrafish as an in vivo model for identification of estrogenic properties of compounds. Live imaging of these transgenic fish provides knowledge of estrogen receptor specificity of different ligands as well as dynamics of estrogen signaling. Coupled to image analysis, the model can provide quantitative dose-response information on estrogenic activity of chemical compounds.
Collapse
Affiliation(s)
- Daniel A Gorelick
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caroline Lucia Pinto
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, 4800 Calhoun Rd., Houston, TX, 77004, USA
| | - Ruixin Hao
- Center for Molecular Toxicology and Carcinogenesis, Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA.,DuPont Haskell Global Centers for Health and Environmental Sciences, Newark, DE, USA
| | - Maria Bondesson
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, 4800 Calhoun Rd., Houston, TX, 77004, USA.
| |
Collapse
|
23
|
Mayrhofer M, Mione M. The Toolbox for Conditional Zebrafish Cancer Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 916:21-59. [PMID: 27165348 DOI: 10.1007/978-3-319-30654-4_2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Here we describe the conditional zebrafish cancer toolbox, which allows for fine control of the expression of oncogenes or downregulation of tumor suppressors at the spatial and temporal level. Methods such as the Gal4/UAS or the Cre/lox systems paved the way to the development of elegant tumor models, which are now being used to study cancer cell biology, clonal evolution, identification of cancer stem cells and anti-cancer drug screening. Combination of these tools, as well as novel developments such as the promising genome editing system through CRISPR/Cas9 and clever application of light reactive proteins will enable the development of even more sophisticated zebrafish cancer models. Here, we introduce this growing toolbox of conditional transgenic approaches, discuss its current application in zebrafish cancer models and provide an outlook on future perspectives.
Collapse
Affiliation(s)
- Marie Mayrhofer
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany
| | - Marina Mione
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
24
|
Yue MS, Plavicki JS, Li XY, Peterson RE, Heideman W. A co-culture assay of embryonic zebrafish hearts to assess migration of epicardial cells in vitro. BMC DEVELOPMENTAL BIOLOGY 2015; 15:50. [PMID: 26715205 PMCID: PMC4696273 DOI: 10.1186/s12861-015-0100-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 12/22/2015] [Indexed: 11/17/2022]
Abstract
Background The vertebrate heart consists of three cell layers: the innermost endothelium, the contractile myocardium and the outermost epicardium. The epicardium is vital for heart development and function, and forms from epicardial progenitor cells (EPCs), which migrate to the myocardium during early development. Disruptions in EPC migration and epicardium formation result in a number of cardiac malformations, many of which resemble congenital heart diseases in humans. Hence, it is important to understand the mechanisms that influence EPC migration and spreading in the developing heart. In vitro approaches heretofore have been limited to monolayer epicardial cell cultures, which may not fully capture the complex interactions that can occur between epicardial and myocardial cells in vivo. Results Here we describe a novel in vitro co-culture assay for assessing epicardial cell migration using embryonic zebrafish hearts. We isolated donor hearts from embryonic zebrafish carrying an epicardial-specific fluorescent reporter after epicardial cells were present on the heart. These were co-cultured with recipient hearts expressing a myocardial-specific fluorescent reporter, isolated prior to EPC migration. Using this method, we can clearly visualize the movement of epicardial cells from the donor heart onto the myocardium of the recipient heart. We demonstrate the utility of this method by showing that epicardial cell migration is significantly delayed or absent when myocardial cells lack contractility and when myocardial cells are deficient in tbx5 expression. Conclusions We present a method to assess the migration of epicardial cells in an in vitro assay, wherein the migration of epicardial cells from a donor heart onto the myocardium of a recipient heart in co-culture is monitored and scored. The donor and recipient hearts can be independently manipulated, using either genetic tools or pharmacological agents. This allows flexibility in experimental design for determining the role that target genes/signaling pathways in specific cell types may have on epicardial cell migration. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0100-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Monica S Yue
- Molecular and Environmental Toxicology Center, University of Wisconsin, 1300 University Avenue, Madison, WI, 53706, USA.
| | - Jessica S Plavicki
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI, 53705, USA.
| | - Xin-yi Li
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi, 710062, China.
| | - Richard E Peterson
- Molecular and Environmental Toxicology Center, University of Wisconsin, 1300 University Avenue, Madison, WI, 53706, USA. .,Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI, 53705, USA.
| | - Warren Heideman
- Molecular and Environmental Toxicology Center, University of Wisconsin, 1300 University Avenue, Madison, WI, 53706, USA. .,Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, 777 Highland Avenue, Madison, WI, 53705, USA.
| |
Collapse
|
25
|
Ramezani T, Laux DW, Bravo IR, Tada M, Feng Y. Live imaging of innate immune and preneoplastic cell interactions using an inducible Gal4/UAS expression system in larval zebrafish skin. J Vis Exp 2015. [PMID: 25741625 PMCID: PMC4354608 DOI: 10.3791/52107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Here we describe a method to conditionally induce epithelial cell transformation by the use of the 4-Hydroxytamoxifen (4-OHT) inducible KalTA4-ERT2/UAS expression system(1) in zebrafish larvae, and the subsequent live imaging of innate immune cell interaction with HRASG12V expressing skin cells. The KalTA4-ERT2/UAS system is both inducible and reversible which allows us to induce cell transformation with precise temporal/spatial resolution in vivo. This provides us with a unique opportunity to live image how individual preneoplastic cells interact with host tissues as soon as they emerge, then follow their progression as well as regression. Recent studies in zebrafish larvae have shown a trophic function of innate immunity in the earliest stages of tumorigenesis(2,3). Our inducible system would allow us to live image the onset of cellular transformation and the subsequent host response, which may lead to important insights on the underlying mechanisms for the regulation of oncogenic trophic inflammatory responses. We also discuss how one might adapt our protocol to achieve temporal and spatial control of ectopic gene expression in any tissue of interest.
Collapse
Affiliation(s)
- Thomas Ramezani
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh
| | - Derek W Laux
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh
| | - Isabel R Bravo
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh
| | - Masazumi Tada
- Department of Cell & Developmental Biology, University College London
| | - Yi Feng
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh;
| |
Collapse
|
26
|
Clay MR, Halloran MC. Cadherin 6 promotes neural crest cell detachment via F-actin regulation and influences active Rho distribution during epithelial-to-mesenchymal transition. Development 2014; 141:2506-15. [PMID: 24917505 DOI: 10.1242/dev.105551] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The epithelial-to-mesenchymal transition (EMT) is a complex change in cell phenotype that is important for cell migration, morphogenesis and carcinoma metastasis. Loss of epithelial cell adhesion and tight regulation of cadherin adhesion proteins are crucial for EMT. Cells undergoing EMT often display cadherin switching, where they downregulate one cadherin and induce expression of another. However, the functions of the upregulated cadherins and their effects on cell motility are poorly understood. Neural crest cells (NCCs), which undergo EMT during development, lose N-cadherin and upregulate Cadherin 6 (Cdh6) prior to EMT. Cdh6 has been suggested to suppress EMT via cell adhesion, but also to promote EMT by mediating pro-EMT signals. Here, we determine novel roles for Cdh6 in generating cell motility during EMT. We use live imaging of NCC behavior in vivo to show that Cdh6 promotes detachment of apical NCC tails, an important early step of EMT. Furthermore, we show that Cdh6 affects spatiotemporal dynamics of F-actin and active Rho GTPase, and that Cdh6 is required for accumulation of F-actin in apical NCC tails during detachment. Moreover, Cdh6 knockdown alters the subcellular distribution of active Rho, which is known to promote localized actomyosin contraction that is crucial for apical NCC detachment. Together, these data suggest that Cdh6 is an important determinant of where subcellular actomyosin forces are generated during EMT. Our results also identify mechanisms by which an upregulated cadherin can generate cell motility during EMT.
Collapse
Affiliation(s)
- Matthew R Clay
- Cell and Molecular Biology Program, University of Wisconsin, Madison, WI 53706, USA Department of Zoology, University of Wisconsin, Madison, WI 53706, USA Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Mary C Halloran
- Cell and Molecular Biology Program, University of Wisconsin, Madison, WI 53706, USA Department of Zoology, University of Wisconsin, Madison, WI 53706, USA Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
27
|
Integration of signals along orthogonal axes of the vertebrate neural tube controls progenitor competence and increases cell diversity. PLoS Biol 2014; 12:e1001907. [PMID: 25026549 PMCID: PMC4098999 DOI: 10.1371/journal.pbio.1001907] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 06/05/2014] [Indexed: 12/21/2022] Open
Abstract
FGF gates competence to generate Floor Plate and Neural Crest in response to Shh and BMP signals by controlling expression of the transcription factor Nkx1.2. A relatively small number of signals are responsible for the variety and pattern of cell types generated in developing embryos. In part this is achieved by exploiting differences in the concentration or duration of signaling to increase cellular diversity. In addition, however, changes in cellular competence—temporal shifts in the response of cells to a signal—contribute to the array of cell types generated. Here we investigate how these two mechanisms are combined in the vertebrate neural tube to increase the range of cell types and deliver spatial control over their location. We provide evidence that FGF signaling emanating from the posterior of the embryo controls a change in competence of neural progenitors to Shh and BMP, the two morphogens that are responsible for patterning the ventral and dorsal regions of the neural tube, respectively. Newly generated neural progenitors are exposed to FGF signaling, and this maintains the expression of the Nk1-class transcription factor Nkx1.2. Ventrally, this acts in combination with the Shh-induced transcription factor FoxA2 to specify floor plate cells and dorsally in combination with BMP signaling to induce neural crest cells. As development progresses, the intersection of FGF with BMP and Shh signals is interrupted by axis elongation, resulting in the loss of Nkx1.2 expression and allowing the induction of ventral and dorsal interneuron progenitors by Shh and BMP signaling to supervene. Hence a similar mechanism increases cell type diversity at both dorsal and ventral poles of the neural tube. Together these data reveal that tissue morphogenesis produces changes in the coincidence of signals acting along orthogonal axes of the neural tube and this is used to define spatial and temporal transitions in the competence of cells to interpret morphogen signaling. During embryonic development different cell types arise at different times and places. This diversity is produced by a relatively small number of signals and depends, at least in part, on changes in the way cells respond to each signal. One example of this so-called change in “competence” is found in the vertebrate spinal cord where a signal, Sonic Hedgehog (Shh), induces a glial cell type known as floor plate (FP) at early developmental times, while the same signal later induces specific types of neurons. Here, we dissected the molecular mechanism underlying the change in competence, and found that another signal, FGF, is involved through its control of the transcription factor Nkx1.2. In embryos, Shh and FGF are produced perpendicular to one another and FP is induced where the two signals intersect. The position of this intersection changes as the embryo elongates and this determines the place and time FP is produced. A similar strategy also appears to apply to another cell type, neural crest. In this case, the intersection of FGF with BMP signal is crucial. Together the data provide new insight into the spatiotemporal control of cell type specification during development of the vertebrate spinal cord.
Collapse
|
28
|
Computational and experimental approaches to reveal the effects of single nucleotide polymorphisms with respect to disease diagnostics. Int J Mol Sci 2014; 15:9670-717. [PMID: 24886813 PMCID: PMC4100115 DOI: 10.3390/ijms15069670] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 12/25/2022] Open
Abstract
DNA mutations are the cause of many human diseases and they are the reason for natural differences among individuals by affecting the structure, function, interactions, and other properties of DNA and expressed proteins. The ability to predict whether a given mutation is disease-causing or harmless is of great importance for the early detection of patients with a high risk of developing a particular disease and would pave the way for personalized medicine and diagnostics. Here we review existing methods and techniques to study and predict the effects of DNA mutations from three different perspectives: in silico, in vitro and in vivo. It is emphasized that the problem is complicated and successful detection of a pathogenic mutation frequently requires a combination of several methods and a knowledge of the biological phenomena associated with the corresponding macromolecules.
Collapse
|
29
|
Akerberg AA, Stewart S, Stankunas K. Spatial and temporal control of transgene expression in zebrafish. PLoS One 2014; 9:e92217. [PMID: 24643048 PMCID: PMC3958484 DOI: 10.1371/journal.pone.0092217] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/19/2014] [Indexed: 01/12/2023] Open
Abstract
Transgenic zebrafish research has provided valuable insights into gene functions and cell behaviors directing vertebrate development, physiology, and disease models. Most approaches use constitutive transgene expression and therefore do not provide control over the timing or levels of transgene induction. We describe an inducible gene expression system that uses new tissue-specific zebrafish transgenic lines that express the Gal4 transcription factor fused to the estrogen-binding domain of the human estrogen receptor. We show these Gal4-ERT driver lines confer rapid, tissue-specific induction of UAS-controlled transgenes following tamoxifen exposure in both embryos and adult fish. We demonstrate how this technology can be used to define developmental windows of gene function by spatiotemporal-controlled expression of constitutively active Notch1 in embryos. Given the array of existing UAS lines, the modular nature of this system will enable many previously intractable zebrafish experiments.
Collapse
Affiliation(s)
- Alexander A Akerberg
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America; Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Scott Stewart
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America
| | - Kryn Stankunas
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States of America; Department of Biology, University of Oregon, Eugene, Oregon, United States of America
| |
Collapse
|
30
|
Calzolari S, Terriente J, Pujades C. Cell segregation in the vertebrate hindbrain relies on actomyosin cables located at the interhombomeric boundaries. EMBO J 2014; 33:686-701. [PMID: 24569501 DOI: 10.1002/embj.201386003] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Segregating cells into compartments during embryonic development is essential for growth and pattern formation. Physical mechanisms shaping compartment boundaries were recently explored in Drosophila, where actomyosin-based barriers were revealed to be important for keeping cells apart. In vertebrates, interhombomeric boundaries are straight interfaces, which often serve as signaling centers that pattern the surrounding tissue. Here, we demonstrate that in the hindbrain of zebrafish embryos cell sorting sharpens the molecular boundaries and, once borders are straight, actomyosin barriers are key to keeping rhombomeric cells segregated. Actomyosin cytoskeletal components are enriched at interhombomeric boundaries, forming cable-like structures in the apical side of the neuroepithelial cells by the time morphological boundaries are visible. When myosin II function is inhibited, cable structures do not form, leading to rhombomeric cell mixing. Downregulation of EphA4a compromises actomyosin cables and cells with different rhombomeric identity intermingle, and the phenotype is rescued enhancing myosin II activity. Moreover, enrichment of actomyosin structures is obtained when EphA4 is ectopically expressed in even-numbered rhombomeres. These findings suggest that mechanical barriers act downstream of EphA/ephrin signaling to segregate cells from different rhombomeres.
Collapse
Affiliation(s)
- Simone Calzolari
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra Parc de Recerca Biomèdica de Barcelona, Barcelona, Spain
| | | | | |
Collapse
|
31
|
Koole W, Tijsterman M. Mosaic analysis and tumor induction in zebrafish by microsatellite instability-mediated stochastic gene expression. Dis Model Mech 2014; 7:929-36. [PMID: 24487406 PMCID: PMC4073281 DOI: 10.1242/dmm.014365] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mosaic analysis, in which two or more populations of cells with differing genotypes are studied in a single animal, is a powerful approach to study developmental mechanisms and gene function in vivo. Over recent years, several genetic methods have been developed to achieve mosaicism in zebrafish, but despite their advances, limitations remain and different approaches and further refinements are warranted. Here, we describe an alternative approach for creating somatic mosaicism in zebrafish that relies on the instability of microsatellite sequences during replication. We placed the coding sequences of various marker proteins downstream of a microsatellite and out-of-frame; in vivo frameshifting into the proper reading frame results in expression of the protein in random individual cells that are surrounded by wild-type cells. We optimized this approach for the binary Gal4-UAS expression system by generating a driver line and effector lines that stochastically express Gal4-VP16 or UAS:H2A-EGFP and self-maintaining UAS:H2A-EGFP-Kaloop, respectively. To demonstrate the utility of this system, we stochastically expressed a constitutively active form of the human oncogene H-RAS and show the occurrence of hyperpigmentation and sporadic tumors within 5 days. Our data demonstrate that inducing somatic mosaicism through microsatellite instability can be a valuable approach for mosaic analysis and tumor induction in Danio rerio.
Collapse
Affiliation(s)
- Wouter Koole
- Department of Toxicogenetics, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
| | - Marcel Tijsterman
- Department of Toxicogenetics, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands.
| |
Collapse
|
32
|
Jacob J, Ribes V, Moore S, Constable SC, Sasai N, Gerety SS, Martin DJ, Sergeant CP, Wilkinson DG, Briscoe J. Valproic acid silencing of ascl1b/Ascl1 results in the failure of serotonergic differentiation in a zebrafish model of fetal valproate syndrome. Dis Model Mech 2013; 7:107-17. [PMID: 24135485 PMCID: PMC3882053 DOI: 10.1242/dmm.013219] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fetal valproate syndrome (FVS) is caused by in utero exposure to the drug sodium valproate. Valproate is used worldwide for the treatment of epilepsy, as a mood stabiliser and for its pain-relieving properties. In addition to birth defects, FVS is associated with an increased risk of autism spectrum disorder (ASD), which is characterised by abnormal behaviours. Valproate perturbs multiple biochemical pathways and alters gene expression through its inhibition of histone deacetylases. Which, if any, of these mechanisms is relevant to the genesis of its behavioural side effects is unclear. Neuroanatomical changes associated with FVS have been reported and, among these, altered serotonergic neuronal differentiation is a consistent finding. Altered serotonin homeostasis is also associated with autism. Here we have used a chemical-genetics approach to investigate the underlying molecular defect in a zebrafish FVS model. Valproate causes the selective failure of zebrafish central serotonin expression. It does so by downregulating the proneural gene ascl1b, an ortholog of mammalian Ascl1, which is a known determinant of serotonergic identity in the mammalian brainstem. ascl1b is sufficient to rescue serotonin expression in valproate-treated embryos. Chemical and genetic blockade of the histone deacetylase Hdac1 downregulates ascl1b, consistent with the Hdac1-mediated silencing of ascl1b expression by valproate. Moreover, tonic Notch signalling is crucial for ascl1b repression by valproate. Concomitant blockade of Notch signalling restores ascl1b expression and serotonin expression in both valproate-exposed and hdac1 mutant embryos. Together, these data provide a molecular explanation for serotonergic defects in FVS and highlight an epigenetic mechanism for genome-environment interaction in disease.
Collapse
Affiliation(s)
- John Jacob
- Division of Developmental Biology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London, NW7 1AA, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Weber T, Köster R. Genetic tools for multicolor imaging in zebrafish larvae. Methods 2013; 62:279-91. [DOI: 10.1016/j.ymeth.2013.07.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 07/08/2013] [Accepted: 07/16/2013] [Indexed: 02/06/2023] Open
|