1
|
Blanc-Legendre M, Guillot L, Chevalier L, Malleret C, Le Menach K, Pardon P, Budzinski H, Brion F, Sire S, Coumailleau P, Charlier TD, Pellegrini E, Cousin X. Long-term impact of embryonic exposure to ethinylestradiol and clotrimazole on behavior and neuroplasticity in zebrafish (Danio rerio). ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2025; 113:104592. [PMID: 39581484 DOI: 10.1016/j.etap.2024.104592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/10/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Estrogen receptors (ER) are widely expressed in the brain of many species and experimental results highlighted the role of estradiol in neuronal plasticity and behavior. Consequently, the brain is therefore a prime target for endocrine disrupting chemicals (EDCs) interacting with estrogen signaling. Very little is known about the late effects of early disruption of estrogen signaling by EDCs. We focused on: ethinylestradiol (EE2; ER agonist) and clotrimazole (inhibitor of key steroidogenesis enzymes, including aromatases). Zebrafish eleutheroembryos were exposed (0-5 days) and then raised normally until adulthood. Several behavioral tests were performed in adults, then cell proliferation and dopaminergic neurons were quantified in several brain regions using immunostaining. Overall, a developmental exposure to EDCs stimulates cell proliferation in the dorsal telencephalon. At environmentally-relevant concentrations, male fish exposed to EE2 exhibited increased activity levels and decreased social behavior, posing a potential risk to population balance and health.
Collapse
Affiliation(s)
- M Blanc-Legendre
- MARBEC, University of Montpellier, CNRS, Ifremer, IRD, INRAE, Palavas-les-Flots, France
| | - L Guillot
- University of Rennes, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S, Rennes 1085, France
| | - L Chevalier
- University of Rennes, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S, Rennes 1085, France
| | - C Malleret
- University of Rennes, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S, Rennes 1085, France
| | - K Le Menach
- University Bordeaux, CNRS, Bordeaux INP, EPOC, UMR 5805, Pessac 33600, France
| | - P Pardon
- University Bordeaux, CNRS, Bordeaux INP, EPOC, UMR 5805, Pessac 33600, France
| | - H Budzinski
- University Bordeaux, CNRS, Bordeaux INP, EPOC, UMR 5805, Pessac 33600, France
| | - F Brion
- Ecotoxicologie des substances et des milieux, Parc ALATA, INERIS, Verneuil-en-Halatte 60550, France
| | - S Sire
- MARBEC, University of Montpellier, CNRS, Ifremer, IRD, INRAE, Palavas-les-Flots, France
| | - P Coumailleau
- University of Rennes, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S, Rennes 1085, France
| | - T D Charlier
- University of Rennes, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S, Rennes 1085, France; University of Rennes, ImPACcell Platform, Biosit, Rennes, France
| | - E Pellegrini
- University of Rennes, Inserm, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S, Rennes 1085, France
| | - X Cousin
- MARBEC, University of Montpellier, CNRS, Ifremer, IRD, INRAE, Palavas-les-Flots, France.
| |
Collapse
|
2
|
Moreno-Oñate M, Gallardo-Fuentes L, Martínez-García PM, Naranjo S, Jiménez-Gancedo S, Tena JJ, Santos-Pereira JM. Rewiring of the epigenome and chromatin architecture by exogenously induced retinoic acid signaling during zebrafish embryonic development. Nucleic Acids Res 2024; 52:3682-3701. [PMID: 38321954 PMCID: PMC11040003 DOI: 10.1093/nar/gkae065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/15/2024] [Accepted: 01/24/2024] [Indexed: 02/08/2024] Open
Abstract
Retinoic acid (RA) is the ligand of RA receptors (RARs), transcription factors that bind to RA response elements. RA signaling is required for multiple processes during embryonic development, including body axis extension, hindbrain antero-posterior patterning and forelimb bud initiation. Although some RA target genes have been identified, little is known about the genome-wide effects of RA signaling during in vivo embryonic development. Here, we stimulate the RA pathway by treating zebrafish embryos with all-trans-RA (atRA) and use a combination of RNA-seq, ATAC-seq, ChIP-seq and HiChIP to gain insight into the molecular mechanisms by which exogenously induced RA signaling controls gene expression. We find that RA signaling is involved in anterior/posterior patterning, central nervous system development, and the transition from pluripotency to differentiation. AtRA treatment also alters chromatin accessibility during early development and promotes chromatin binding of RARαa and the RA targets Hoxb1b, Meis2b and Sox3, which cooperate in central nervous system development. Finally, we show that exogenous RA induces a rewiring of chromatin architecture, with alterations in chromatin 3D interactions involving target genes. Altogether, our findings identify genome-wide targets of RA signaling and provide a molecular mechanism by which developmental signaling pathways regulate target gene expression by altering chromatin topology.
Collapse
Affiliation(s)
- Marta Moreno-Oñate
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Lourdes Gallardo-Fuentes
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Pedro M Martínez-García
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Silvia Naranjo
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Sandra Jiménez-Gancedo
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - Juan J Tena
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas/Universidad Pablo de Olavide, 41013 Sevilla, Spain
| | - José M Santos-Pereira
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Biología Celular, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain
| |
Collapse
|
3
|
Umeda K, Tanaka K, Chowdhury G, Nasu K, Kuroyanagi Y, Yamasu K. Evolutionarily conserved roles of foxg1a in the developing subpallium of zebrafish embryos. Dev Growth Differ 2024; 66:219-234. [PMID: 38378191 PMCID: PMC11457518 DOI: 10.1111/dgd.12917] [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: 10/31/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/22/2024]
Abstract
The vertebrate telencephalic lobes consist of the pallium (dorsal) and subpallium (ventral). The subpallium gives rise to the basal ganglia, encompassing the pallidum and striatum. The development of this region is believed to depend on Foxg1/Foxg1a functions in both mice and zebrafish. This study aims to elucidate the genetic regulatory network controlled by foxg1a in subpallium development using zebrafish as a model. The expression gradient of foxg1a within the developing telencephalon was examined semi-quantitatively in initial investigations. Utilizing the CRISPR/Cas9 technique, we subsequently established a foxg1a mutant line and observed the resultant phenotypes. Morphological assessment revealed that foxg1a mutants exhibit a thin telencephalon together with a misshapen preoptic area (POA). Notably, accumulation of apoptotic cells was identified in this region. In mutants at 24 h postfertilization, the expression of pallium markers expanded ventrally, while that of subpallium markers was markedly suppressed. Concurrently, the expression of fgf8a, vax2, and six3b was shifted ventrally, causing anomalous expression in regions typical of POA formation in wild-type embryos. Consequently, the foxg1a mutation led to expansion of the pallium and disrupted the subpallium and POA. This highlights a pivotal role of foxg1a in directing the dorsoventral patterning of the telencephalon, particularly in subpallium differentiation, mirroring observations in mice. Additionally, reduced expression of neural progenitor maintenance genes was detected in mutants, suggesting the necessity of foxg1a in preserving neural progenitors. Collectively, these findings underscore evolutionarily conserved functions of foxg1 in the development of the subpallium in vertebrate embryos.
Collapse
Affiliation(s)
- Koto Umeda
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Kaiho Tanaka
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Gazlima Chowdhury
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
- Department of Aquatic Environment and Resource ManagementSher‐e‐Bangla Agricultural UniversityDhakaBangladesh
| | - Kouhei Nasu
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Yuri Kuroyanagi
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| |
Collapse
|
4
|
Minařík M, Modrell MS, Gillis JA, Campbell AS, Fuller I, Lyne R, Micklem G, Gela D, Pšenička M, Baker CVH. Identification of multiple transcription factor genes potentially involved in the development of electrosensory versus mechanosensory lateral line organs. Front Cell Dev Biol 2024; 12:1327924. [PMID: 38562141 PMCID: PMC10982350 DOI: 10.3389/fcell.2024.1327924] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
In electroreceptive jawed vertebrates, embryonic lateral line placodes give rise to electrosensory ampullary organs as well as mechanosensory neuromasts. Previous reports of shared gene expression suggest that conserved mechanisms underlie electroreceptor and mechanosensory hair cell development and that electroreceptors evolved as a transcriptionally related "sister cell type" to hair cells. We previously identified only one transcription factor gene, Neurod4, as ampullary organ-restricted in the developing lateral line system of a chondrostean ray-finned fish, the Mississippi paddlefish (Polyodon spathula). The other 16 transcription factor genes we previously validated in paddlefish were expressed in both ampullary organs and neuromasts. Here, we used our published lateral line organ-enriched gene-set (arising from differential bulk RNA-seq in late-larval paddlefish), together with a candidate gene approach, to identify 25 transcription factor genes expressed in the developing lateral line system of a more experimentally tractable chondrostean, the sterlet (Acipenser ruthenus, a small sturgeon), and/or that of paddlefish. Thirteen are expressed in both ampullary organs and neuromasts, consistent with conservation of molecular mechanisms. Seven are electrosensory-restricted on the head (Irx5, Irx3, Insm1, Sp5, Satb2, Mafa and Rorc), and five are the first-reported mechanosensory-restricted transcription factor genes (Foxg1, Sox8, Isl1, Hmx2 and Rorb). However, as previously reported, Sox8 is expressed in ampullary organs as well as neuromasts in a catshark (Scyliorhinus canicula), suggesting the existence of lineage-specific differences between cartilaginous and ray-finned fishes. Overall, our results support the hypothesis that ampullary organs and neuromasts develop via largely conserved transcriptional mechanisms, and identify multiple transcription factors potentially involved in the formation of electrosensory versus mechanosensory lateral line organs.
Collapse
Affiliation(s)
- Martin Minařík
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Melinda S. Modrell
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - J. Andrew Gillis
- Department of Zoology, University of Cambridge, Cambridge, United Kingdom
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA, United States
| | - Alexander S. Campbell
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Isobel Fuller
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Rachel Lyne
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Gos Micklem
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - David Gela
- Faculty of Fisheries and Protection of Waters, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Vodňany, Czechia
| | - Martin Pšenička
- Faculty of Fisheries and Protection of Waters, Research Institute of Fish Culture and Hydrobiology, University of South Bohemia in České Budějovice, Vodňany, Czechia
| | - Clare V. H. Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
5
|
Chowdhury G, Umeda K, Ohyanagi T, Nasu K, Yamasu K. Involvement of nr2f genes in brain regionalization and eye development during early zebrafish development. Dev Growth Differ 2024; 66:145-160. [PMID: 38263801 PMCID: PMC11457503 DOI: 10.1111/dgd.12912] [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: 11/27/2023] [Revised: 01/02/2024] [Accepted: 01/06/2024] [Indexed: 01/25/2024]
Abstract
Nuclear receptor subfamily 2 group F (Nr2f) proteins are essential for brain development in mice, but little is known about their precise roles and their evolutionary diversification. In the present study, the expression patterns of major nr2f genes (nr2f1a, nr2f1b, and nr2f2) during early brain development were investigated in zebrafish. Comparisons of their expression patterns revealed similar but temporally and spatially distinct patterns after early somite stages in the brain. Frameshift mutations in the three nr2f genes, achieved using the CRISPR/Cas9 method, resulted in a smaller telencephalon and smaller eyes in the nr2f1a mutants; milder forms of those defects were present in the nr2f1b and nr2f2 mutants. Acridine orange staining revealed enhanced cell death in the brain and/or eyes in all nr2f homozygous mutants. The expression of regional markers in the brain did not suggest global defects in brain regionalization; however, shha expression in the preoptic area and hypothalamus, as well as fgf8a expression in the anterior telencephalon, was disturbed in nr2f1a and nr2f1b mutants, potentially leading to a defective telencephalon. Specification of the retina and optic stalk was also significantly affected. The overexpression of nr2f1b by injection of mRNA disrupted the anterior brain at a high dose, and the expression of pax6a in the eyes and fgf8a in the telencephalon at a low dose. The results of these loss- and gain-of-function approaches showed that nr2f genes regulate the development of the telencephalon and eyes in zebrafish embryos.
Collapse
Affiliation(s)
- Gazlima Chowdhury
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
- Department of Aquatic Environment and Resource ManagementSher‐e‐Bangla Agricultural UniversityDhakaBangladesh
| | - Koto Umeda
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Takero Ohyanagi
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Kouhei Nasu
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| | - Kyo Yamasu
- Division of Life Science, Graduate School of Science and EngineeringSaitama UniversitySaitamaJapan
| |
Collapse
|
6
|
Geng Y, Zou H, Guo Y, Huang M, Wu Y, Hou L. Chronic exposure to cortisone induces thyroid endocrine disruption and retinal dysfunction in adult female zebrafish (Danio rerio). THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 905:167022. [PMID: 37709101 DOI: 10.1016/j.scitotenv.2023.167022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 09/09/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023]
Abstract
Cortisone has a large content in rivers because of its wide range of medical applications and elimination by organisms that naturally secrete it. As a steroid hormone, cortisone is recognized as a novel endocrine disruptor. Although ecotoxicological effects of the reproductive endocrine system have mainly been reported recently, thyroid endocrine in fish remains relatively less understood. Here, adult female zebrafish were exposed to cortisone at 0.0 (control), 3.2, 38.7, and 326.9 ng/L for 60 days. Evidence in this study came from fish behavior, hormone levels, gene expression, histological and morphological examinations. The results showed that THs (thyroid hormone) level disruption and pathohistological changes occurred in the thyroid gland, which may account for the gene expression changes in the hypothalamus-pituitary-thyroid gland axis. Specifically, more conversion of T4 (thyroxine) to T3 (triiodothyronine) led to an increased TSH (thyroid stimulating hormone) level in plasma. Severe thyroid tissue damage mainly occurred in the zebrafish exposed to 326.9 ng/L of cortisone. Meanwhile, consistent with the THs trend, the fish locomotion activity displayed more anxiety and excitement, the partial blockage of GABA (γ - aminobutyric acid) synthetic pathway genes might be the explanation of the underlying mechanism. Cortisone affected the gene expressions in the visual cycle and the circadian rhythm network also suggested interactions between thyroid endocrine disruption, retinal dysfunction, and abnormal behaviors of zebrafish. In summary, these findings suggest chronic exposure to cortisone induced various adverse effects in adult female zebrafish, which may help us better understand the risk of cortisone to fish in the wild.
Collapse
Affiliation(s)
- Yuxin Geng
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Hong Zou
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Yanfang Guo
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Manlin Huang
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Yashi Wu
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - Liping Hou
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China.
| |
Collapse
|
7
|
Papadogiannis V, Hockman D, Mercurio S, Ramsay C, Hintze M, Patthey C, Streit A, Shimeld SM. Evolution of the expression and regulation of the nuclear hormone receptor ERR gene family in the chordate lineage. Dev Biol 2023; 504:12-24. [PMID: 37696353 DOI: 10.1016/j.ydbio.2023.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 09/01/2023] [Accepted: 09/09/2023] [Indexed: 09/13/2023]
Abstract
The Estrogen Related Receptor (ERR) nuclear hormone receptor genes have a wide diversity of roles in vertebrate development. In embryos, ERR genes are expressed in several tissues, including the central and peripheral nervous systems. Here we seek to establish the evolutionary history of chordate ERR genes, their expression and their regulation. We examine ERR expression in mollusc, amphioxus and sea squirt embryos, finding the single ERR orthologue is expressed in the nervous system in all three, with muscle expression also found in the two chordates. We show that most jawed vertebrates and lampreys have four ERR paralogues, and that vertebrate ERR genes were ancestrally linked to Estrogen Receptor genes. One of the lamprey paralogues shares conserved expression domains with jawed vertebrate ERRγ in the embryonic vestibuloacoustic ganglion, eye, brain and spinal cord. Hypothesising that conserved expression derives from conserved regulation, we identify a suite of pan-vertebrate conserved non-coding sequences in ERR introns. We use transgenesis in lamprey and chicken embryos to show that these sequences are regulatory and drive reporter gene expression in the nervous system. Our data suggest an ancient association between ERR and the nervous system, including expression in cells associated with photosensation and mechanosensation. This includes the origin in the vertebrate common ancestor of a suite of regulatory elements in the 3' introns that drove nervous system expression and have been conserved from this point onwards.
Collapse
Affiliation(s)
| | - Dorit Hockman
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Neuroscience Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Silvia Mercurio
- Department of Environmental Science and Policy, Università Degli Studi di Milano, Via Celoria 2, 20133, Milano, Italy
| | - Claire Ramsay
- Department of Biology, University of Oxford, 11a Mansfield Road, Oxford, OX1 3SZ, UK
| | - Mark Hintze
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Cedric Patthey
- Department of Radiosciences, Umeå University, 901 85, Umeå, Sweden
| | - Andrea Streit
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Sebastian M Shimeld
- Department of Biology, University of Oxford, 11a Mansfield Road, Oxford, OX1 3SZ, UK.
| |
Collapse
|
8
|
Nikaido M, Shirai A, Mizumaki Y, Shigenobu S, Ueno N, Hatta K. Intestinal expression patterns of transcription factors and markers for interstitial cells in the larval zebrafish. Dev Growth Differ 2023; 65:418-428. [PMID: 37452633 DOI: 10.1111/dgd.12878] [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: 04/11/2023] [Revised: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
For the digestion of food, it is important for the gut to be differentiated regionally and to have proper motor control. However, the number of transcription factors that regulate its development is still limited. Meanwhile, the interstitial cells of the gastrointestinal (GI) tract are necessary for intestinal motility in addition to the enteric nervous system. There are anoctamine1 (Ano1)-positive and platelet-derived growth factor receptor α (Pdgfra)-positive interstitial cells in mammal, but Pdgfra-positive cells have not been reported in the zebrafish. To identify new transcription factors involved in GI tract development, we used RNA sequencing comparing between larval and adult gut. We isolated 40 transcription factors that were more highly expressed in the larval gut. We demonstrated expression patterns of the 13 genes, 7 of which were newly found to be expressed in the zebrafish larval gut. Six of the 13 genes encode nuclear receptors. The osr2 is expressed in the anterior part, while foxP4 in its distal part. Also, we reported the expression pattern of pdgfra for the first time in the larval zebrafish gut. Our data provide fundamental knowledge for studying vertebrate gut regionalization and motility by live imaging using zebrafish.
Collapse
Affiliation(s)
| | - Ayaka Shirai
- School of Science, University of Hyogo, Ako-gun, Japan
| | | | - Shuji Shigenobu
- Trans-Scale Biology Center, National Institute for Basic Biology, Okazaki, Japan
| | - Naoto Ueno
- Trans-Scale Biology Center, National Institute for Basic Biology, Okazaki, Japan
- Unit of Quantitative and Imaging Biology, International Research Collaboration Center, National Institute of Natural Sciences, Okazaki, Japan
| | - Kohei Hatta
- Graduate School of Science, University of Hyogo, Ako-gun, Japan
| |
Collapse
|
9
|
Pannetier P, Poulsen R, Gölz L, Coordes S, Stegeman H, Koegst J, Reger L, Braunbeck T, Hansen M, Baumann L. Reversibility of Thyroid Hormone System-Disrupting Effects on Eye and Thyroid Follicle Development in Zebrafish (Danio rerio) Embryos. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2023; 42:1276-1292. [PMID: 36920003 DOI: 10.1002/etc.5608] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 03/10/2023] [Indexed: 05/27/2023]
Abstract
Early vertebrate development is partially regulated by thyroid hormones (THs). Environmental pollutants that interact with the TH system (TH system-disrupting chemicals [THSDCs]) can have massively disrupting effects on this essential phase. Eye development of fish is directly regulated by THs and can, therefore, be used as a thyroid-related endpoint in endocrine disruptor testing. To evaluate the effects of THSDC-induced eye malformations during early development, zebrafish (Danio rerio) embryos were exposed for 5 days postfertilization (dpf) to either propylthiouracil, a TH synthesis inhibitor, or tetrabromobisphenol A, which interacts with TH receptors. Subsequently, one half of the embryos were exposed further to the THSDCs until 8 dpf, while the other half of the embryos were raised in clean water for 3 days to check for reversibility of effects. Continued THSDC exposure altered eye size and pigmentation and induced changes in the cellular structure of the retina. This correlated with morphological alterations of thyroid follicles as revealed by use of a transgenic zebrafish line. Interestingly, effects were partly reversible after a recovery period as short as 3 days. Results are consistent with changes in TH levels measured in different tissues of the embryos, for example, in the eyes. The results show that eye development in zebrafish embryos is very sensitive to THSDC treatment but able to recover quickly from early exposure by effective repair mechanisms. Environ Toxicol Chem 2023;42:1276-1292. © 2023 The Authors. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Pauline Pannetier
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Rikke Poulsen
- Environmental Metabolomics Laboratory, Department of Environmental Science, University of Aarhus, Aarhus, Denmark
| | - Lisa Gölz
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Sara Coordes
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Hanna Stegeman
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Johannes Koegst
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Luisa Reger
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Thomas Braunbeck
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
| | - Martin Hansen
- Environmental Metabolomics Laboratory, Department of Environmental Science, University of Aarhus, Aarhus, Denmark
| | - Lisa Baumann
- Aquatic Ecology & Toxicology, Center for Organismal Studies, University of Heidelberg, Heidelberg, Germany
- Amsterdam Institute for Life and Environment (A-LIFE), Section on Environmental Health & Toxicology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
10
|
Passos LS, Jacinavicius FR, Geraldes V, Nunes de Freitas PN, Helena da Silva G, Costa de Almeida É, Priscila do Carmo Alves A, Orlando TM, da Silva Cerozi B, Teodoro Martinez DS, Pinto E. Ecotoxicological assessment of guanitoxin-producing cyanobacteria in Danio rerio and Daphnia similis. CHEMOSPHERE 2023; 332:138846. [PMID: 37146772 DOI: 10.1016/j.chemosphere.2023.138846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/27/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Anthropogenic activity has dramatically deteriorated aquatic ecosystems in recent years. Such environmental alterations could change the primary producers' composition, exacerbating the proliferation of harmful microorganisms such as cyanobacteria. Cyanobacteria can produce several secondary metabolites, including guanitoxin, a potent neurotoxin and the only naturally occurring anticholinesterase organophosphate ever reported in the literature. Therefore, this study investigated the acute toxicity of guanitoxin-producing cyanobacteria Sphaerospermopsis torques-reginae (ITEP-024 strain) aqueous and 50% methanolic extracts in zebrafish (Danio rerio) hepatocytes (ZF-L cell line), zebrafish embryos (fish embryo toxicity - FET) and specimens of the microcrustacean Daphnia similis. For this, hepatocytes were exposed to 1-500 mg/L of the ITEP-024 extracts for 24 h, the embryos to 31.25-500 mg/L for 96 h, and D. similis to 10-3000 mg/L for 48 h. Non-target metabolomics was also performed to analyze secondary metabolites produced by the ITEP-024 using LC-MS/MS. Metabolomics indicated the guanitoxin presence just in the aqueous extract of the ITEP-024 and the presence of the cyanopeptides namalides, spumigins, and anabaenopeptins in the methanolic extract. The aqueous extract decreased the viability of zebrafish hepatocytes (EC(I)50(24h) = 366.46 mg/L), and the methanolic extract was not toxic. FET showed that the aqueous extract (LC50(96) = 353.55 mg/L) was more toxic than the methanolic extract (LC50(96) = 617.91 mg/L). However, the methanolic extract had more sublethal effects, such as abdominal and cardiac (cardiotoxicity) edema and deformation (spinal curvature of the larvae). Both extracts immobilized daphnids at the highest concentration analyzed. However, the aqueous extract was nine times more lethal (EC(I)50(48h) = 108.2 mg/L) than the methanolic extract (EC(I)50(48h) = 980.65 mg/L). Our results showed an imminent biological risk for aquatic fauna living in an ecosystem surrounded by ITEP-024 metabolites. Our findings thus highlight the urgency of understanding the effects of guanitoxin and cyanopeptides in aquatic animals.
Collapse
Affiliation(s)
- Larissa Souza Passos
- Laboratory of Environmental Biogeochemistry, Center for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, Brazil; Laboratory of Toxins and Natural Algae Products, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Fernanda Rios Jacinavicius
- Laboratory of Toxins and Natural Algae Products, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Vanessa Geraldes
- Laboratory of Environmental Biogeochemistry, Center for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, Brazil; Laboratory of Toxins and Natural Algae Products, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Paloma Nathane Nunes de Freitas
- Laboratory of Environmental Biogeochemistry, Center for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, Brazil
| | - Gabriela Helena da Silva
- National Nanotechnology Laboratory, National Center for Research in Energy and Materials, Campinas, São Paulo, Brazil
| | - Éryka Costa de Almeida
- Laboratory of Toxins and Natural Algae Products, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Tamira Maria Orlando
- Department of Animal Science, "Luiz de Queiroz" College of Agriculture, University of São Paulo, Piracicaba, Brazil
| | - Brunno da Silva Cerozi
- Department of Animal Science, "Luiz de Queiroz" College of Agriculture, University of São Paulo, Piracicaba, Brazil
| | | | - Ernani Pinto
- Laboratory of Environmental Biogeochemistry, Center for Nuclear Energy in Agriculture, University of São Paulo, Piracicaba, Brazil; Laboratory of Toxins and Natural Algae Products, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil; Food Research Center (FoRC - CEPID), University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
11
|
Lazcano I, Pech-Pool SM, Olvera A, García-Martínez I, Palacios-Pérez S, Orozco A. The importance of thyroid hormone signaling during early development: Lessons from the zebrafish model. Gen Comp Endocrinol 2023; 334:114225. [PMID: 36709002 DOI: 10.1016/j.ygcen.2023.114225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
The zebrafish is an optimal experimental model to study thyroid hormone (TH) involvement in vertebrate development. The use of state-of-the-art zebrafish genetic tools available for the study of the effect of gene silencing, cell fate decisions and cell lineage differentiation have contributed to a more insightful comprehension of molecular, cellular, and tissue-specific TH actions. In contrast to intrauterine development, extrauterine embryogenesis observed in zebrafish has facilitated a more detailed study of the development of the hypothalamic-pituitary-thyroid axis. This model has also enabled a more insightful analysis of TH molecular actions upon the organization and function of the brain, the retina, the heart, and the immune system. Consequently, zebrafish has become a trendy model to address paradigms of TH-related functional and biomedical importance. We here compilate the available knowledge regarding zebrafish developmental events for which specific components of TH signaling are essential.
Collapse
Affiliation(s)
- I Lazcano
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - S M Pech-Pool
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - A Olvera
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - I García-Martínez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - S Palacios-Pérez
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico
| | - A Orozco
- Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Boulevard Juriquilla 3001, Campus Juriquilla, Querétaro 76230, Mexico; Escuela Nacional de Estudios Superiores, Unidad Juriquilla, Universidad Nacional Autónoma de México (UNAM), Campus Juriquilla, Querétaro 76230, Mexico.
| |
Collapse
|
12
|
Chen HJ, Barske L, Talbot JC, Dinwoodie OM, Roberts RR, Farmer DT, Jimenez C, Merrill AE, Tucker AS, Crump JG. Nuclear receptor Nr5a2 promotes diverse connective tissue fates in the jaw. Dev Cell 2023; 58:461-473.e7. [PMID: 36905926 PMCID: PMC10050118 DOI: 10.1016/j.devcel.2023.02.011] [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: 09/15/2022] [Revised: 01/06/2023] [Accepted: 02/17/2023] [Indexed: 03/12/2023]
Abstract
Organ development involves the sustained production of diverse cell types with spatiotemporal precision. In the vertebrate jaw, neural-crest-derived progenitors produce not only skeletal tissues but also later-forming tendons and salivary glands. Here we identify the pluripotency factor Nr5a2 as essential for cell-fate decisions in the jaw. In zebrafish and mice, we observe transient expression of Nr5a2 in a subset of mandibular postmigratory neural-crest-derived cells. In zebrafish nr5a2 mutants, nr5a2-expressing cells that would normally form tendons generate excess jaw cartilage. In mice, neural-crest-specific Nr5a2 loss results in analogous skeletal and tendon defects in the jaw and middle ear, as well as salivary gland loss. Single-cell profiling shows that Nr5a2, distinct from its roles in pluripotency, promotes jaw-specific chromatin accessibility and gene expression that is essential for tendon and gland fates. Thus, repurposing of Nr5a2 promotes connective tissue fates to generate the full repertoire of derivatives required for jaw and middle ear function.
Collapse
Affiliation(s)
- Hung-Jhen Chen
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Lindsey Barske
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jared C Talbot
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA
| | - Olivia M Dinwoodie
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - Ryan R Roberts
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Biomedical Sciences, Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - D'Juan T Farmer
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Molecular, Cell and Developmental Biology Department and Orthopaedic Surgery, University of California, Los Angeles, CA 90095, USA
| | - Christian Jimenez
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Amy E Merrill
- Department of Biomedical Sciences, Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA; Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Abigail S Tucker
- Centre for Craniofacial and Regenerative Biology, King's College London, London, SE1 9RT, UK
| | - J Gage Crump
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
13
|
Kraft M, Gölz L, Rinderknecht M, Koegst J, Braunbeck T, Baumann L. Developmental exposure to triclosan and benzophenone-2 causes morphological alterations in zebrafish (Danio rerio) thyroid follicles and eyes. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:33711-33724. [PMID: 36495432 PMCID: PMC9736712 DOI: 10.1007/s11356-022-24531-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/28/2022] [Indexed: 05/31/2023]
Abstract
Thyroid hormones (THs) regulate a multitude of developmental and metabolic processes, which are responsible for vertebrate development, growth, and maintenance of homeostasis. THs also play a key role in neurogenesis of vertebrates and thus affect eye development, which is vital for foraging efficiency and for effective escape from predation. Currently, there are no validated test guidelines for the assessment of TH system-disrupting chemicals (THSDCs) in fish. Consequently, the present study was designed to demonstrate the suitability of novel thyroid-related endpoints in early life-stages of fish. Embryos of a transgenic zebrafish (Danio rerio) line expressing the reporter gene tg:mCherry in their thyrocytes were used to investigate the effects of the environmental THSDCs triclosan (TCS, antibacterial agent) and benzophenone-2 (BP-2, UV filter) on thyroid follicle and eye development. Both BP-2 and TCS caused thyroid follicle hyperplasia in transgenic zebrafish, thus confirming their role as THSDCs. The effect intensity on follicle size and fluorescence was comparable with a 1.7-fold increase for BP-2 and 1.6-fold for TCS. Alterations of the cellular structures of the retina indicate an impact of both substances on eye development, with a stronger impact of TCS. With respect to guideline development, results provide further evidence for the suitability of morphological changes in thyroid follicles and the eyes as novel endpoints for the sensitive assessment of THSD-related effects in fish.
Collapse
Affiliation(s)
- Maximilian Kraft
- Aquatic Toxicology and Ecology Section, Centre of Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
| | - Lisa Gölz
- Aquatic Toxicology and Ecology Section, Centre of Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
| | - Maximilian Rinderknecht
- Aquatic Toxicology and Ecology Section, Centre of Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
| | - Johannes Koegst
- Aquatic Toxicology and Ecology Section, Centre of Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
| | - Thomas Braunbeck
- Aquatic Toxicology and Ecology Section, Centre of Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany
| | - Lisa Baumann
- Aquatic Toxicology and Ecology Section, Centre of Organismal Studies, University of Heidelberg, Im Neuenheimer Feld 504, 69120, Heidelberg, Germany.
| |
Collapse
|
14
|
Concha ML, Reig G. Origin, form and function of extraembryonic structures in teleost fishes. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210264. [PMID: 36252221 PMCID: PMC9574637 DOI: 10.1098/rstb.2021.0264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/12/2022] [Indexed: 12/18/2022] Open
Abstract
Teleost eggs have evolved a highly derived early developmental pattern within vertebrates as a result of the meroblastic cleavage pattern, giving rise to a polar stratified architecture containing a large acellular yolk and a small cellular blastoderm on top. Besides the acellular yolk, the teleost-specific yolk syncytial layer (YSL) and the superficial epithelial enveloping layer are recognized as extraembryonic structures that play critical roles throughout embryonic development. They provide enriched microenvironments in which molecular feedback loops, cellular interactions and mechanical signals emerge to sculpt, among other things, embryonic patterning along the dorsoventral and left-right axes, mesendodermal specification and the execution of morphogenetic movements in the early embryo and during organogenesis. An emerging concept points to a critical role of extraembryonic structures in reinforcing early genetic and morphogenetic programmes in reciprocal coordination with the embryonic blastoderm, providing the necessary boundary conditions for development to proceed. In addition, the role of the enveloping cell layer in providing mechanical, osmotic and immunological protection during early stages of development, and the autonomous nutritional support provided by the yolk and YSL, have probably been key aspects that have enabled the massive radiation of teleosts to colonize every ecological niche on the Earth. This article is part of the theme issue 'Extraembryonic tissues: exploring concepts, definitions and functions across the animal kingdom'.
Collapse
Affiliation(s)
- Miguel L. Concha
- Integrative Biology Program, Institute of Biomedical Sciences (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Biomedical Neuroscience Institute (BNI), Santiago 8380453, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 7800003, Chile
| | - Germán Reig
- Escuela de Tecnología Médica y del Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 7800003, Chile
| |
Collapse
|
15
|
Li X, Liu S, Qi D, Qi H, Wang Y, Zhao K, Tian F. Genome-wide identification and expression of the peroxisome proliferator-activated receptor gene family in the Tibetan highland fish Gymnocypris przewalskii. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:1685-1699. [PMID: 36469183 DOI: 10.1007/s10695-022-01152-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) plays an important role in the regulation of lipid metabolism and has been widely identified in diverse species. Gymnocypris przewalskii is a native fish of the Qinghai Tibetan Plateau that survives in a chronically cold environment. In the current study, we conducted genome-wide identification of PPAR genes, revealing the existence of seven PPARs in the G. przewalskii genome. Collinearity was observed between two copies of PPARαb and PPARγ in G. przewalskii, suggesting that the additional copy might be gained through whole genome duplication. Both phylogenetic and multiple sequence alignment analyses indicated that PPARs in G. przewalskii were conserved with teleosts. The cold treatment (10 °C and 4 °C) led to the developmental delay of G. przewalskii embryos. Continuous expression of PPARs was observed during the embryonic development of G. przewalskii under normal and cold conditions, with significantly different transcriptional patterns. These results indicated that PPARs participated in the embryonic development of G. przewalskii, and were involved in the cold response during development. The current study proposed a potential role of PPARs in the cold response in the embryonic development of G. przewalskii, which shed light on understanding cold adaptation in Tibetan highland fish.
Collapse
Affiliation(s)
- Xiaohuan Li
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, No. 23 Xinning Road, Xining, 810001, Qinghai, China
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sijia Liu
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, No. 23 Xinning Road, Xining, 810001, Qinghai, China
| | - Delin Qi
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, Qinghai, China
| | - Hongfang Qi
- Qinghai Provincial Key Laboratory of Breeding and Protection of Gymnocypris Przewalskii, Xining, Qinghai, China
| | - Yang Wang
- Qinghai Provincial Key Laboratory of Breeding and Protection of Gymnocypris Przewalskii, Xining, Qinghai, China
| | - Kai Zhao
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, No. 23 Xinning Road, Xining, 810001, Qinghai, China.
| | - Fei Tian
- Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, No. 23 Xinning Road, Xining, 810001, Qinghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
16
|
Heppert JK, Lickwar CR, Tillman MC, Davis BR, Davison JM, Lu HY, Chen W, Busch-Nentwich EM, Corcoran DL, Rawls JF. Conserved roles for Hnf4 family transcription factors in zebrafish development and intestinal function. Genetics 2022; 222:iyac133. [PMID: 36218393 PMCID: PMC9713462 DOI: 10.1093/genetics/iyac133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 12/13/2022] Open
Abstract
Transcription factors play important roles in the development of the intestinal epithelium and its ability to respond to endocrine, nutritional, and microbial signals. Hepatocyte nuclear factor 4 family nuclear receptors are liganded transcription factors that are critical for the development and function of multiple digestive organs in vertebrates, including the intestinal epithelium. Zebrafish have 3 hepatocyte nuclear factor 4 homologs, of which, hnf4a was previously shown to mediate intestinal responses to microbiota in zebrafish larvae. To discern the functions of other hepatocyte nuclear factor 4 family members in zebrafish development and intestinal function, we created and characterized mutations in hnf4g and hnf4b. We addressed the possibility of genetic redundancy amongst these factors by creating double and triple mutants which showed different rates of survival, including apparent early lethality in hnf4a; hnf4b double mutants and triple mutants. RNA sequencing performed on digestive tracts from single and double mutant larvae revealed extensive changes in intestinal gene expression in hnf4a mutants that were amplified in hnf4a; hnf4g mutants, but limited in hnf4g mutants. Changes in hnf4a and hnf4a; hnf4g mutants were reminiscent of those seen in mice including decreased expression of genes involved in intestinal function and increased expression of cell proliferation genes, and were validated using transgenic reporters and EdU labeling in the intestinal epithelium. Gnotobiotics combined with RNA sequencing also showed hnf4g has subtler roles than hnf4a in host responses to microbiota. Overall, phenotypic changes in hnf4a single mutants were strongly enhanced in hnf4a; hnf4g double mutants, suggesting a conserved partial genetic redundancy between hnf4a and hnf4g in the vertebrate intestine.
Collapse
Affiliation(s)
- Jennifer K Heppert
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Colin R Lickwar
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA
| | - Matthew C Tillman
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA
| | - Briana R Davis
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA
| | - James M Davison
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA
| | - Hsiu-Yi Lu
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA
| | - Wei Chen
- Center for Genomics and Computational Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - David L Corcoran
- Center for Genomics and Computational Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
17
|
Gómez-Abellán V, Pérez-Oliva AB, Cabas I, Hermi F, Arizcun M, García-Moreno D, Sepulcre MP, Mulero V. Peroxisome proliferator-activated receptors alpha and beta mediate the anti-inflammatory effects of the cyclopentenone prostaglandin 15-deoxy-Δ 12,14-PGJ 2 in fish granulocytes. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 136:104498. [PMID: 35948178 DOI: 10.1016/j.dci.2022.104498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
Prostaglandins (PGs) are highly reactive small lipophilic molecules derived from polyunsaturated fatty acids of the cell membrane and play a key role in the resolution of inflammation processes. 15-deoxy-Δ12,14-PGJ2 (15dPGJ2) is a cyclopentenone PG (CyPG) of the J series with anti-inflammatory, anti-proliferative and pro-apoptotic effects. This CyPG can signal through: (i) the PGD2 receptor (DP2) and peroxisome proliferator-activated receptor γ (PPARγ) or (ii) by covalent binding to protein nucleophiles, such as, thiols groups of cysteine, lysine or histidine via a Michael addition reaction, modifying its structure and function. In this work we show that acidophilic granulocytes (AGs) of gilthead seabream (Sparus aurata L.), the functional equivalent to mammalian neutrophils, constitutively expressed ppara, pparb and pparg genes, the latter showing the highest expression and up-regulation when stimulated by bacterial DNA. In addition, we tested the ability of 15dPGJ2, and its biotinylated analog, as well as several PPARγ ligands, to modulate reactive oxygen species (ROS) and/or cytokines production during a Toll like receptor (TLR)-mediated granulocyte response. Thus, 15dPGJ2 was able to significantly decrease bacterial DNA-induced ROS production and transcript levels of pparg, interleukin-1β (il1b) and prostaglandin-endoperoxide synthase 2 (ptgs2). In contrast, its biotinylated analog was less potent and a higher dose was required to elicit the same effects on ROS production and cytokine expression. In addition, different PPARγ agonists were able to mimic the effects of 15dPGJ2. Conversely, the PPARγ antagonist T007097 abolished the effect of 15dPGJ2 on DNA bacterial-induced ROS production. Surprisingly, transactivation assays revealed that both 15dPGJ2 and its biotinylated analog signaled via Pparα and Pparβ, but not by Pparγ. These results were further confirmed by HPLC/MS analysis, where Pparβ was identified as an interactor of biotin-15dPGJ2 in naïve and DNA-stimulated leukocytes. Taken together, our data show that 15dPGJ2 acts both through Ppar activation and covalent binding to proteins in fish granulocytes and identify for the first time in vertebrates a role for Pparα and Pparβ in the resolution of inflammation mediated by 15dPGJ2.
Collapse
Affiliation(s)
- Victoria Gómez-Abellán
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain
| | - Ana B Pérez-Oliva
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Isabel Cabas
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Fatma Hermi
- Unit of Immunology, Environmental Microbiology and Cancerously, Faculty of Sciences of Bizerte, Jarzouna, Bizerte, 7021, University of Carthage, Tunis, Tunisia
| | - Marta Arizcun
- Oceanographic Center of Murcia, Spanish Institute of Oceanography (IEO-CSIC), 30860, Murcia, Spain
| | - Diana García-Moreno
- Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - María P Sepulcre
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Victoriano Mulero
- Departamento de Biología Cellular e Histología, Facultad de Biología, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, 30120, Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
18
|
Xu H, Su J, Ku T, Liu QS, Liang J, Ren Z, Zhou Q, Jiang G. Constructing an MCF-7 breast cancer cell-based transient transfection assay for screening RARα (Ant)agonistic activities of emerging phenolic compounds. JOURNAL OF HAZARDOUS MATERIALS 2022; 435:129024. [PMID: 35523094 DOI: 10.1016/j.jhazmat.2022.129024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
The screening of compounds with endocrine disrupting effects has been attracting increasing attention due to the continuous release of emerging chemicals into the environment. Testing the (ant)agonistic activities of these chemicals on the retinoic acid receptor α (RARα), a vital nuclear receptor, is necessary to explain their perturbation in the endocrine system in vivo. In the present study, MCF-7 breast carcinoma cells were transiently transfected with a RARα expression vector (pEF1α-RARα-RFP) and a reporter vector containing a retinoic acid reaction element (pRARE-TA-Luc). Under optimized conditions, the performance of the newly constructed system was evaluated for its feasibility in screening the (ant)agonistic effects of emerging phenolic compounds on RARα. The results showed that this transient transfection cell model responded well to stimulation with (ant)agonists of RARα, and the EC50 and IC50 values were 0.87 nM and 2.67 μM for AM580 and Ro41-5253, respectively. Its application in testing several emerging phenolic compounds revealed that triclosan (TCS) and tetrabromobisphenol A (TBBPA) exerted notable RARα antagonistic activities. This newly developed bioassay based on MCF-7 is promising in identifying the agonistic or antagonistic activities of xenobiotics on RARα and has good potential for studying RARα signaling-involved toxicological effects of emerging chemicals of concern.
Collapse
Affiliation(s)
- Hanqing Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; National and Local Joint Engineering Research Center of Ecological Treatment Technology for Urban Water Pollution, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Su
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingting Ku
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Environment and Resources, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Jiefeng Liang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhihua Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Environment and Health, Jianghan University, Wuhan 430056, China.
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
19
|
Søderstrøm S, Lille-Langøy R, Yadetie F, Rauch M, Milinski A, Dejaegere A, Stote RH, Goksøyr A, Karlsen OA. Agonistic and potentiating effects of perfluoroalkyl substances (PFAS) on the Atlantic cod (Gadus morhua) peroxisome proliferator-activated receptors (Ppars). ENVIRONMENT INTERNATIONAL 2022; 163:107203. [PMID: 35364415 DOI: 10.1016/j.envint.2022.107203] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
Toxicity mediated by per- and polyfluoroalkyl substances (PFAS), and especially perfluoroalkyl acids (PFAAs), has been linked to activation of peroxisome proliferator-activated receptors (Ppar) in many vertebrates. Here, we present the primary structures, phylogeny, and tissue-specific distributions of the Atlantic cod (Gadus morhua) gmPpara1, gmPpara2, gmPparb, and gmPparg, and demonstrate that the carboxylic acids PFHxA, PFOA, PFNA, as well as the sulfonic acid PFHxS, activate gmPpara1 in vitro, which was also supported by in silico analyses. Intriguingly, a binary mixture of PFOA and the non-activating PFOS produced a higher activation of gmPpara1 compared to PFOA alone, suggesting that PFOS has a potentiating effect on receptor activation. Supporting the experimental data, docking and molecular dynamics simulations of single and double-ligand complexes led to the identification of a putative allosteric binding site, which upon binding of PFOS stabilizes an active conformation of gmPpara1. Notably, binary exposures of gmPpara1, gmPpara2, and gmPparb to model-agonists and PFAAs produced similar potentiating effects. This study provides novel mechanistic insights into how PFAAs may modulate the Ppar signaling pathway by either binding the canonical ligand-binding pocket or by interacting with an allosteric binding site. Thus, individual PFAAs, or mixtures, could potentially modulate the Ppar-signaling pathway in Atlantic cod by interfering with at least one gmPpar subtype.
Collapse
Affiliation(s)
- Sofie Søderstrøm
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway; Institute of Marine Research, Nordnesgaten 50, NO-5005 Bergen, Norway(1)
| | - Roger Lille-Langøy
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway; Institute of Marine Research, Nordnesgaten 50, NO-5005 Bergen, Norway(1)
| | - Fekadu Yadetie
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway
| | - Mateusz Rauch
- Department of Integrated Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, Illkirch, France
| | - Ana Milinski
- Department of Integrated Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, Illkirch, France
| | - Annick Dejaegere
- Department of Integrated Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, Illkirch, France
| | - Roland H Stote
- Department of Integrated Structural Biology, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Institut National de La Santé et de La Recherche Médicale (INSERM), U1258/Centre National de Recherche Scientifique (CNRS), UMR7104/Université de Strasbourg, Illkirch, France
| | - Anders Goksøyr
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway
| | - Odd André Karlsen
- Department of Biological Sciences, University of Bergen, Thormøhlens gate 53 A/B, NO-5006 Bergen, Norway.
| |
Collapse
|
20
|
Machado AM, Gomes-dos-Santos A, Fonseca MM, da Fonseca RR, Veríssimo A, Felício M, Capela R, Alves N, Santos M, Salvador-Caramelo F, Domingues M, Ruivo R, Froufe E, Castro LFC. A genome assembly of the Atlantic chub mackerel ( Scomber colias): a valuable teleost fishing resource. GIGABYTE 2022; 2022:gigabyte40. [PMID: 36824513 PMCID: PMC9650269 DOI: 10.46471/gigabyte.40] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/09/2022] [Indexed: 11/09/2022] Open
Abstract
The Atlantic chub mackerel, Scomber colias (Gmelin, 1789), is a medium-sized pelagic fish with substantial importance in the fisheries of the Atlantic Ocean and the Mediterranean Sea. Over the past decade, this species has gained special relevance, being one of the main targets of pelagic fisheries in the NE Atlantic. Here, we sequenced and annotated the first high-quality draft genome assembly of S. colias, produced with PacBio HiFi long reads and Illumina paired-end short reads. The estimated genome size is 814 Mbp, distributed into 2,028 scaffolds and 2,093 contigs with an N50 length of 4.19 and 3.34 Mbp, respectively. We annotated 27,675 protein-coding genes and the BUSCO analyses indicated high completeness, with 97.3% of the single-copy orthologs in the Actinopterygii library profile. The present genome assembly represents a valuable resource to address the biology and management of this relevant fishery. Finally, this genome assembly ranks fourth in high-quality genome assemblies within the order Scombriformes and first in the genus Scomber.
Collapse
Affiliation(s)
- André M. Machado
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, U. Porto - University of Porto, Portugal
| | - André Gomes-dos-Santos
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, U. Porto - University of Porto, Portugal
| | - Miguel M. Fonseca
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
| | - Rute R. da Fonseca
- Center for Global Mountain Biodiversity, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Center for Macroecology, Evolution, and Climate, GLOBE Institute, University of Copenhagen, Denmark
| | - Ana Veríssimo
- CIBIO - Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO - Laboratório Associado, Campus de Vairão, Universidade do Porto, 4485-661 Vairão, Portugal
- BIOPOLIS - Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, 4485-661 Vairão, Portugal
| | - Mónica Felício
- Portuguese Institute for the Sea and Atmosphere, I.P. (IPMA), Portugal
| | - Ricardo Capela
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, U. Porto - University of Porto, Portugal
| | - Nélson Alves
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, U. Porto - University of Porto, Portugal
| | - Miguel Santos
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, U. Porto - University of Porto, Portugal
| | - Filipe Salvador-Caramelo
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, U. Porto - University of Porto, Portugal
| | - Marcos Domingues
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
| | - Raquel Ruivo
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
| | - Elsa Froufe
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
| | - L. Filipe C. Castro
- CIIMAR – Interdisciplinary Centre of Marine and Environmental Research, U. Porto – University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, U. Porto - University of Porto, Portugal
| |
Collapse
|
21
|
The Preservation of PPARγ Genome Duplicates in Some Teleost Lineages: Insights into Lipid Metabolism and Xenobiotic Exploitation. Genes (Basel) 2022; 13:genes13010107. [PMID: 35052447 PMCID: PMC8774674 DOI: 10.3390/genes13010107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/28/2021] [Accepted: 12/31/2021] [Indexed: 01/10/2023] Open
Abstract
Three peroxisome proliferator-activated receptor paralogues (PPARα, -β and -γ) are currently recognized in vertebrate genomes. PPARγ is known to modulate nutrition, adipogenesis and immunity in vertebrates. Natural ligands of PPARγ have been proposed; however, the receptor also binds synthetic ligands such as endocrine disruptors. Two paralogues of PPARα and PPARβ have been documented in teleost species, a consequence of the 3R WGD. Recently, two PPARγ paralogue genes were also identified in Astyanax mexicanus. We aimed to determine whether the presence of two PPARγ paralogues is prevalent in other teleost genomes, through genomic and phylogenetic analysis. Our results showed that besides Characiformes, two PPARγ paralogous genes were also identified in other teleost taxa, coinciding with the teleost-specific, whole-genome duplication and with the retention of both genes prior to the separation of the Clupeocephala. To functionally characterize these genes, we used the European sardine (Sardina pilchardus) as a model. PPARγA and PPARγB display a different tissue distribution, despite the similarity of their functional profiles: they are unresponsive to tested fatty acids and other human PPARγ ligands yet yield a transcriptional response in the presence of tributyltin (TBT). This observation puts forward the relevance of comparative analysis to decipher alternative binding architectures and broadens the disruptive potential of man-made chemicals for aquatic species.
Collapse
|
22
|
Takesono A, Kudoh T, Tyler CR. Application of Transgenic Zebrafish Models for Studying the Effects of Estrogenic Endocrine Disrupting Chemicals on Embryonic Brain Development. Front Pharmacol 2022; 13:718072. [PMID: 35264948 PMCID: PMC8900011 DOI: 10.3389/fphar.2022.718072] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 01/14/2022] [Indexed: 11/13/2022] Open
Abstract
Endocrine disrupting chemicals (EDCs) are environmental pollutants that mimic hormones and/or disrupt their function. Estrogenic EDCs (eEDCs) interfere with endogenous estrogen signalling pathway(s) and laboratory animal and human epidemiological studies have provided evidence for a causal link between exposure to them during embryonic/early life and neurological impairments. However, our understanding of the molecular and cellular mechanism(s) underlying eEDCs exposure effects on brain development, tissue architecture and function and behaviour are limited. Transgenic (TG) zebrafish models offer new approach methodologies (NAMs) to help identify the modes of action (MoAs) of EDCs and their associated impacts on tissue development and function. Estrogen biosensor TG zebrafish models have been applied to study eEDC interactions and resulting transcriptional activation (via a fluorescent reporter expression) across the entire body of the developing zebrafish embryo, including in real time. These estrogen biosensor TG zebrafish models are starting to deepen our understanding of the spatiotemporal actions of eEDCs and their resulting impacts on neurological development, brain function and behaviour. In this review, we first investigate the links between early life exposure to eEDCs and neurodevelopmental alterations in model organisms (rodents and zebrafish) and humans. We then present examples of the application of estrogen biosensor and other TG zebrafish models for elucidating the mechanism(s) underlying neurodevelopmental toxicities of eEDCs. In particular we illustrate the utility of combining estrogen biosensor zebrafish models with other TG zebrafish models for understanding the effects of eEDCs on the brain, spanning cellular processes, brain circuitry, neurophysiology and behaviour. Finally, we discuss the future prospects of TG zebrafish models as experimental models for studying more complex scenarios for exposure to contaminant mixtures on neurological development and function.
Collapse
Affiliation(s)
- Aya Takesono
- *Correspondence: Aya Takesono, ; Charles R. Tyler,
| | | | | |
Collapse
|
23
|
Coppola U, Waxman JS. Origin and evolutionary landscape of Nr2f transcription factors across Metazoa. PLoS One 2021; 16:e0254282. [PMID: 34807940 PMCID: PMC8608329 DOI: 10.1371/journal.pone.0254282] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/07/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Nuclear Receptor Subfamily 2 Group F (Nr2f) orphan nuclear hormone transcription factors (TFs) are fundamental regulators of many developmental processes in invertebrates and vertebrates. Despite the importance of these TFs throughout metazoan development, previous work has not clearly outlined their evolutionary history. RESULTS We integrated molecular phylogeny with comparisons of intron/exon structure, domain architecture, and syntenic conservation to define critical evolutionary events that distinguish the Nr2f gene family in Metazoa. Our data indicate that a single ancestral eumetazoan Nr2f gene predated six main Bilateria subfamilies, which include single Nr2f homologs, here referred to as Nr2f1/2/5/6, that are present in invertebrate protostomes and deuterostomes, Nr2f1/2 homologs in agnathans, and Nr2f1, Nr2f2, Nr2f5, and Nr2f6 orthologs that are found in gnathostomes. Four cnidarian Nr2f1/2/5/6 and three agnathan Nr2f1/2 members are each due to independent expansions, while the vertebrate Nr2f1/Nr2f2 and Nr2f5/Nr2f6 members each form paralogous groups that arose from the established series of whole-genome duplications (WGDs). Nr2f6 members are the most divergent Nr2f subfamily in gnathostomes. Interestingly, in contrast to the other gnathostome Nr2f subfamilies, Nr2f5 has been independently lost in numerous vertebrate lineages. Furthermore, our analysis shows there are differential expansions and losses of Nr2f genes in teleosts following their additional rounds of WGDs. CONCLUSION Overall, our analysis of Nr2f gene evolution helps to reveal the origins and previously unrecognized relationships of this ancient TF family, which may allow for greater insights into the conservation of Nr2f functions that shape Metazoan body plans.
Collapse
Affiliation(s)
- Ugo Coppola
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, United States of America
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Research Foundation, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| |
Collapse
|
24
|
Neural is Fundamental: Neural Stemness as the Ground State of Cell Tumorigenicity and Differentiation Potential. Stem Cell Rev Rep 2021; 18:37-55. [PMID: 34714532 DOI: 10.1007/s12015-021-10275-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2021] [Indexed: 01/07/2023]
Abstract
Tumorigenic cells are similar to neural stem cells or embryonic neural cells in regulatory networks, tumorigenicity and pluripotent differentiation potential. By integrating the evidence from developmental biology, tumor biology and evolution, I will make a detailed discussion on the observations and propose that neural stemness underlies two coupled cell properties, tumorigenicity and pluripotent differentiation potential. Neural stemness property of tumorigenic cells can hopefully integrate different observations/concepts underlying tumorigenesis. Neural stem cells and tumorigenic cells share regulatory networks; both exhibit neural stemness, tumorigenicity and pluripotent differentiation potential; both depend on expression or activation of ancestral genes; both rely primarily on aerobic glycolytic metabolism; both can differentiate into various cells/tissues that are derived from three germ layers, leading to tumor formation resembling severely disorganized or more degenerated process of embryonic tissue differentiation; both are enriched in long genes with more splice variants that provide more plastic scaffolds for cell differentiation, etc. Neural regulatory networks, which include higher levels of basic machineries of cell physiological functions and developmental programs, work concertedly to define a basic state with fast cell cycle and proliferation. This is predestined by the evolutionary advantage of neural state, the ground or initial state for multicellularity with adaptation to an ancient environment. Tumorigenesis might represent a process of restoration of neural ground state, thereby restoring a state with fast proliferation and pluripotent differentiation potential in somatic cells. Tumorigenesis and pluripotent differentiation potential might be better understood from understanding neural stemness, and cancer therapy should benefit more from targeting neural stemness.
Collapse
|
25
|
Coral JA, Heaps S, Glaholt SP, Karty JA, Jacobson SC, Shaw JR, Bondesson M. Arsenic exposure induces a bimodal toxicity response in zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117637. [PMID: 34182391 DOI: 10.1016/j.envpol.2021.117637] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 05/25/2023]
Abstract
In toxicology, standard sigmoidal concentration-response curves are used to predict effects concentrations and set chemical regulations. However, current literature also establishes the existence of complex, bimodal concentration-response curves, as is the case for arsenic toxicity. This bimodal response has been observed at the molecular level, but not characterized at the whole organism level. This study investigated the effect of arsenic (sodium arsenite) on post-gastrulated zebrafish embryos and elucidated effects of bimodal concentration-responses on different phenotypic perturbations. Six hour post fertilized (hpf) zebrafish embryos were exposed to arsenic to 96 hpf. Hatching success, mortality, and morphometric endpoints were evaluated both in embryos with chorions and dechorionated embryos. Zebrafish embryos exhibited a bimodal response to arsenic exposure. Concentration-response curves for exposed embryos with intact chorions had an initial peak in mortality (88%) at 1.33 mM arsenic, followed by a decrease in toxicity (~20% mortality) at 1.75 mM, and subsequently peaked to 100% mortality at higher concentrations. To account for the bimodal response, two distinct concentration-response curves were generated with estimated LC10 values (and 95% CI) of 0.462 (0.415, 0.508) mM and 1.69 (1.58, 1.78) mM for the 'low concentration' and 'high concentration' peaks, respectively. Other phenotypic analyses, including embryo length, yolk and pericardial edema all produced similar concentration-response patterns. Tests with dechorionated embryos also resulted in a bimodal toxicity response but with lower LC10 values of 0.170 (0.120, 0.220) mM and 0.800 (0.60, 0842) mM, respectively. Similarities in bimodal concentration-responses between with-chorion and dechorionated embryos indicate that the observed effect was not caused by the chorion limiting arsenic availability, thus lending support to other studies such as those that hypothesized a conserved bimodal mechanism of arsenic interference with nuclear receptor activation.
Collapse
Affiliation(s)
- Jason A Coral
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA.
| | - Samuel Heaps
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Stephen P Glaholt
- O'Neill School of Public and Environmental Affairs, Indiana University, Bloomington, IN, USA
| | - Jonathan A Karty
- Department of Chemistry, Indiana University, Bloomington, IN, USA
| | | | - Joseph R Shaw
- O'Neill School of Public and Environmental Affairs, Indiana University, Bloomington, IN, USA
| | - Maria Bondesson
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| |
Collapse
|
26
|
Eide M, Zhang X, Karlsen OA, Goldstone JV, Stegeman J, Jonassen I, Goksøyr A. The chemical defensome of five model teleost fish. Sci Rep 2021; 11:10546. [PMID: 34006915 PMCID: PMC8131381 DOI: 10.1038/s41598-021-89948-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 05/04/2021] [Indexed: 12/13/2022] Open
Abstract
How an organism copes with chemicals is largely determined by the genes and proteins that collectively function to defend against, detoxify and eliminate chemical stressors. This integrative network includes receptors and transcription factors, biotransformation enzymes, transporters, antioxidants, and metal- and heat-responsive genes, and is collectively known as the chemical defensome. Teleost fish is the largest group of vertebrate species and can provide valuable insights into the evolution and functional diversity of defensome genes. We have previously shown that the xenosensing pregnane x receptor (pxr, nr1i2) is lost in many teleost species, including Atlantic cod (Gadus morhua) and three-spined stickleback (Gasterosteus aculeatus), but it is not known if compensatory mechanisms or signaling pathways have evolved in its absence. In this study, we compared the genes comprising the chemical defensome of five fish species that span the teleosteii evolutionary branch often used as model species in toxicological studies and environmental monitoring programs: zebrafish (Danio rerio), medaka (Oryzias latipes), Atlantic killifish (Fundulus heteroclitus), Atlantic cod, and three-spined stickleback. Genome mining revealed evolved differences in the number and composition of defensome genes that can have implication for how these species sense and respond to environmental pollutants, but we did not observe any candidates of compensatory mechanisms or pathways in cod and stickleback in the absence of pxr. The results indicate that knowledge regarding the diversity and function of the defensome will be important for toxicological testing and risk assessment studies.
Collapse
Affiliation(s)
- Marta Eide
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Xiaokang Zhang
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital-Radiumhospitalet, Oslo, Norway
| | - Odd André Karlsen
- Department of Biological Sciences, University of Bergen, Bergen, Norway
| | - Jared V Goldstone
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - John Stegeman
- Biology Department, Woods Hole Oceanographic Institution, Woods Hole, MA, USA
| | - Inge Jonassen
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Anders Goksøyr
- Department of Biological Sciences, University of Bergen, Bergen, Norway.
| |
Collapse
|
27
|
Farnsworth DR, Posner M, Miller AC. Single cell transcriptomics of the developing zebrafish lens and identification of putative controllers of lens development. Exp Eye Res 2021; 206:108535. [PMID: 33705730 PMCID: PMC8092445 DOI: 10.1016/j.exer.2021.108535] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/31/2021] [Accepted: 03/02/2021] [Indexed: 01/10/2023]
Abstract
The vertebrate lens is a valuable model system for investigating the gene expression changes that coordinate tissue differentiation due to its inclusion of two spatially separated cell types, the outer epithelial cells and the deeper denucleated fiber cells that they support. Zebrafish are a useful model system for studying lens development given the organ's rapid development in the first several days of life in an accessible, transparent embryo. While we have strong foundational knowledge of the diverse lens crystallin proteins and the basic gene regulatory networks controlling lens development, no study has detailed gene expression in a vertebrate lens at single cell resolution. Here we report an atlas of lens gene expression in zebrafish embryos and larvae at single cell resolution through five days of development, identifying a number of novel putative regulators of lens development. Our data address open questions about the temperospatial expression of α-crystallins during lens development that will support future studies of their function and provide the first detailed view of β- and γ-crystallin expression in and outside the lens. We describe divergent expression in transcription factor genes that occur as paralog pairs in the zebrafish. Finally, we examine the expression dynamics of cytoskeletal, membrane associated, RNA-binding, and transcription factor genes, identifying a number of novel patterns. Overall these data provide a foundation for identifying and characterizing lens developmental regulatory mechanisms and revealing targets for future functional studies with potential therapeutic impact.
Collapse
Affiliation(s)
| | - Mason Posner
- Department of Biology and Toxicology, Ashland University, Ashland, OH, USA.
| | - Adam C Miller
- Institute of Neuroscience, University of Oregon, Eugene, OR, USA
| |
Collapse
|
28
|
Chambers JM, Addiego A, Flores-Mireles AL, Wingert RA. Ppargc1a Controls Ciliated Cell Development by Regulating Prostaglandin Biosynthesis. Cell Rep 2020; 33:108370. [PMID: 33176142 PMCID: PMC7731726 DOI: 10.1016/j.celrep.2020.108370] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 07/23/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022] Open
Abstract
Cilia are microtubule-based organelles that function in a multitude of physiological contexts to perform chemosensing, mechanosensing, and fluid propulsion. The process of ciliogenesis is highly regulated, and disruptions result in disease states termed ciliopathies. Here, we report that peroxisome proliferator-activated receptor gamma, coactivator 1 alpha (ppargc1a) is essential for ciliogenesis in nodal, mono-, and multiciliated cells (MCCs) and for discernment of renal tubule ciliated cell fate during embryogenesis. ppargc1a performs these functions by affecting prostaglandin signaling, whereby cilia formation and renal MCC fate are restored with prostaglandin E2 (PGE2) treatment in ppargc1a-deficient animals. Genetic disruption of ppargc1a specifically reduces expression of the prostanoid biosynthesis gene prostaglandin-endoperoxide synthase 1 (ptgs1), and suboptimal knockdown of both genes shows this synergistic effect. Furthermore, ptgs1 overexpression rescues ciliogenesis and renal MCCs in ppargc1a-deficient embryos. These findings position Ppargc1a as a key genetic regulator of prostaglandin signaling during ciliated cell ontogeny.
Collapse
Affiliation(s)
- Joseph M Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Amanda Addiego
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Ana L Flores-Mireles
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, 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, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
29
|
Beato S, Toledo-Solís FJ, Fernández I. Vitamin K in Vertebrates' Reproduction: Further Puzzling Pieces of Evidence from Teleost Fish Species. Biomolecules 2020; 10:E1303. [PMID: 32917043 PMCID: PMC7564532 DOI: 10.3390/biom10091303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
Vitamin K (VK) is a fat-soluble vitamin that vertebrates have to acquire from the diet, since they are not able to de novo synthesize it. VK has been historically known to be required for the control of blood coagulation, and more recently, bone development and homeostasis. Our understanding of the VK metabolism and the VK-related molecular pathways has been also increased, and the two main VK-related pathways-the pregnane X receptor (PXR) transactivation and the co-factor role on the γ-glutamyl carboxylation of the VK dependent proteins-have been thoroughly investigated during the last decades. Although several studies evidenced how VK may have a broader VK biological function than previously thought, including the reproduction, little is known about the specific molecular pathways. In vertebrates, sex differentiation and gametogenesis are tightly regulated processes through a highly complex molecular, cellular and tissue crosstalk. Here, VK metabolism and related pathways, as well as how gametogenesis might be impacted by VK nutritional status, will be reviewed. Critical knowledge gaps and future perspectives on how the different VK-related pathways come into play on vertebrate's reproduction will be identified and proposed. The present review will pave the research progress to warrant a successful reproductive status through VK nutritional interventions as well as towards the establishment of reliable biomarkers for determining proper nutritional VK status in vertebrates.
Collapse
Affiliation(s)
- Silvia Beato
- Campus de Vegazana, s/n, Universidad de León (ULE), 24071 León, Spain;
| | - Francisco Javier Toledo-Solís
- Consejo Nacional de Ciencia y Tecnología (CONACYT, México), Av. Insurgentes Sur 1582, Col. Crédito Constructor, Alcaldía Benito Juárez, C.P. 03940 Ciudad de Mexico, Mexico;
- Department of Biology and Geology, University of Almería, 04120 Almería, Spain
| | - Ignacio Fernández
- Center for Aquaculture Research, Agrarian Technological Institute of Castile and Leon, Ctra. Arévalo, s/n, 40196 Zamarramala, Segovia, Spain
| |
Collapse
|
30
|
Parsons AE, Lange A, Hutchinson TH, Miyagawa S, Iguchi T, Kudoh T, Tyler CR. Expression dynamics of genes in the hypothalamic-pituitary-thyroid (HPT) cascade and their responses to 3,3',5-triiodo-l-thyronine (T3) highlights potential vulnerability to thyroid-disrupting chemicals in zebrafish (Danio rerio) embryo-larvae. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2020; 225:105547. [PMID: 32623180 DOI: 10.1016/j.aquatox.2020.105547] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 06/08/2020] [Accepted: 06/14/2020] [Indexed: 06/11/2023]
Abstract
Some chemicals in the environment disrupt thyroid hormone (TH) systems leading to alterations in organism development, but their effect mechanisms are poorly understood. In fish, this has been limited by a lack of fundamental knowledge on thyroid gene ontogeny and tissue expression in early life stages. Here we established detailed expression profiles for a suite of genes in the hypothalamic-pituitary-thyroid (HPT) axis of zebrafish (Danio rerio) between 24-120 h post fertilisation (hpf) and quantified their responses following exposure to 3,3',5-triiodo-L-thyronine (T3) using whole mount in situ hybridisation (WISH) and qRT-PCR (using whole-body extracts). All of the selected genes in the HPT axis demonstrated dynamic transcript expression profiles across the developmental stages examined. The expression of thyroid receptor alpha (thraa) was observed in the brain, gastrointestinal tract, craniofacial tissues and pectoral fins, while thyroid receptor beta (thrb) expression occurred in the brain, otic vesicles, liver and lower jaw. The TH deiodinases (dio1, dio2 and dio3b) were expressed in the liver, pronephric ducts and brain and the patterns differed depending on life stage. Both dio1 and dio2 were also expressed in the intestinal bulb (96-120 hpf), and dio2 expression occurred also in the pituitary (48-120 hpf). Exposure of zebrafish embryo-larvae to T3 (30 and 100 μg L-1) for periods of 48, 96 or 120 hpf resulted in the up-regulation of thraa, thrb, dio3b, thyroid follicle synthesis proteins (pax8) and corticotropin-releasing hormone (crhb) and down-regulation of dio1, dio2, glucuronidation enzymes (ugt1ab) and thyroid stimulating hormone (tshb) (assessed via qRT-PCR) and responses differed across life stage and tissues. T3 induced thraa expression in the pineal gland, pectoral fins, brain, somites, gastrointestinal tract, craniofacial tissues, liver and pronephric ducts. T3 enhanced thrb expression in the brain, jaw cartilage and intestine, while thrb expression was suppressed in the liver. T3 exposure suppressed the transcript levels of dio1 and dio2 in the liver, brain, gastrointestinal tract and craniofacial tissues, while dio2 signalling was also suppressed in the pituitary gland. Dio3b expression was induced by T3 exposure in the jaw cartilage, pectoral fins and brain. The involvement of THs in the development of numerous body tissues and the responsiveness of these tissues to T3 in zebrafish highlights their potential vulnerability to exposure to environmental thyroid-disrupting chemicals.
Collapse
Affiliation(s)
- Aoife E Parsons
- University of Exeter, Biosciences, College of Life and Environmental Sciences, Geoffrey Pope Building, Stocker Rd., Exeter, EX4 4QD, United Kingdom
| | - Anke Lange
- University of Exeter, Biosciences, College of Life and Environmental Sciences, Geoffrey Pope Building, Stocker Rd., Exeter, EX4 4QD, United Kingdom
| | - Thomas H Hutchinson
- University of Plymouth, School of Geography, Earth & Environmental Sciences, Drake Circus, Plymouth, Devon, PL4 8AA, United Kingdom
| | - Shinichi Miyagawa
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, 125-8585, Japan
| | - Taisen Iguchi
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Kanagawa, 236-0027, Japan
| | - Tetsuhiro Kudoh
- University of Exeter, Biosciences, College of Life and Environmental Sciences, Geoffrey Pope Building, Stocker Rd., Exeter, EX4 4QD, United Kingdom
| | - Charles R Tyler
- University of Exeter, Biosciences, College of Life and Environmental Sciences, Geoffrey Pope Building, Stocker Rd., Exeter, EX4 4QD, United Kingdom.
| |
Collapse
|
31
|
Abstract
As the first organ to form and function in all vertebrates, the heart is crucial to development. Tightly-regulated levels of retinoic acid (RA) are critical for the establishment of the regulatory networks that drive normal cardiac development. Thus, the heart is an ideal organ to investigate RA signaling, with much work remaining to be done in this area. Herein, we highlight the role of RA signaling in vertebrate heart development and provide an overview of the field's inception, its current state, and in what directions it might progress so that it may yield fruitful insight for therapeutic applications within the domain of regenerative medicine.
Collapse
|
32
|
Santoriello C, Sporrij A, Yang S, Flynn RA, Henriques T, Dorjsuren B, Custo Greig E, McCall W, Stanhope ME, Fazio M, Superdock M, Lichtig A, Adatto I, Abraham BJ, Kalocsay M, Jurynec M, Zhou Y, Adelman K, Calo E, Zon LI. RNA helicase DDX21 mediates nucleotide stress responses in neural crest and melanoma cells. Nat Cell Biol 2020; 22:372-379. [PMID: 32231306 PMCID: PMC7185069 DOI: 10.1038/s41556-020-0493-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 02/24/2020] [Indexed: 12/25/2022]
Abstract
The availability of nucleotides has a direct impact on transcription. The inhibition of dihydroorotate dehydrogenase (DHODH) with leflunomide impacts nucleotide pools by reducing pyrimidine levels. Leflunomide abrogates the effective transcription elongation of genes required for neural crest development and melanoma growth in vivo1. To define the mechanism of action, we undertook an in vivo chemical suppressor screen for restoration of neural crest after leflunomide treatment. Surprisingly, we found that alterations in progesterone and progesterone receptor (Pgr) signalling strongly suppressed leflunomide-mediated neural crest effects in zebrafish. In addition, progesterone bypasses the transcriptional elongation block resulting from Paf complex deficiency, rescuing neural crest defects in ctr9 morphant and paf1(alnz24) mutant embryos. Using proteomics, we found that Pgr binds the RNA helicase protein Ddx21. ddx21-deficient zebrafish show resistance to leflunomide-induced stress. At a molecular level, nucleotide depletion reduced the chromatin occupancy of DDX21 in human A375 melanoma cells. Nucleotide supplementation reversed the gene expression signature and DDX21 occupancy changes prompted by leflunomide. Together, our results show that DDX21 acts as a sensor and mediator of transcription during nucleotide stress.
Collapse
Affiliation(s)
- Cristina Santoriello
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Audrey Sporrij
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Song Yang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Ryan A Flynn
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Telmo Henriques
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Bilguujin Dorjsuren
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Eugenia Custo Greig
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Wyatt McCall
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Meredith E Stanhope
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Maurizio Fazio
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Michael Superdock
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Asher Lichtig
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Isaac Adatto
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Brian J Abraham
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marian Kalocsay
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Michael Jurynec
- Department of Orthopedics, University of Utah, Salt Lake City, UT, USA
| | - Yi Zhou
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA
| | - Karen Adelman
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Eliezer Calo
- Department of Biology and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Leonard I Zon
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA. .,Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Harvard Stem Cell Institute, Howard Hughes Medical Institute and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
33
|
Fonseca ESS, Hiromori Y, Kaite Y, Ruivo R, Franco JN, Nakanishi T, Santos MM, Castro LFC. An Orthologue of the Retinoic Acid Receptor (RAR) Is Present in the Ecdysozoa Phylum Priapulida. Genes (Basel) 2019; 10:genes10120985. [PMID: 31795452 PMCID: PMC6947571 DOI: 10.3390/genes10120985] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/26/2019] [Accepted: 11/27/2019] [Indexed: 12/19/2022] Open
Abstract
Signalling molecules and their cognate receptors are central components of the Metazoa endocrine system. Defining their presence or absence in extant animal lineages is critical to accurately devise evolutionary patterns, physiological shifts and the impact of endocrine disrupting chemicals. Here, we address the evolution of retinoic acid (RA) signalling in the Priapulida worm, Priapulus caudatus Lamarck, 1816, an Ecdysozoa. RA signalling has been shown to be central to chordate endocrine homeostasis, participating in multiple developmental and physiological processes. Priapulids, with their slow rate of molecular evolution and phylogenetic position, represent a key taxon to investigate the early phases of Ecdysozoa evolution. By exploring a draft genome assembly, we show, by means of phylogenetics and functional assays, that an orthologue of the nuclear receptor retinoic acid receptor (RAR) subfamily, a central mediator of RA signalling, is present in Ecdysozoa, contrary to previous perception. We further demonstrate that the Priapulida RAR displays low-affinity for retinoids (similar to annelids), and is not responsive to common endocrine disruptors acting via RAR. Our findings provide a timeline for RA signalling evolution in the Bilateria and give support to the hypothesis that the increase in RA affinity towards RAR is a late acquisition in the evolution of the Metazoa.
Collapse
Affiliation(s)
- Elza S. S. Fonseca
- CIIMAR/CIMAR Interdisciplinary Centre of Marine and Environmental Research, U.Porto, 4450-208 Matosinhos, Portugal; (E.S.S.F.); (R.R.); (J.N.F.)
- FCUP—Faculty of Sciences, Department of Biology, U.Porto, 4169-007 Porto, Portugal
| | - Youhei Hiromori
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; (Y.H.); (Y.K.)
- Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka 513-8670, Japan
| | - Yoshifumi Kaite
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; (Y.H.); (Y.K.)
| | - Raquel Ruivo
- CIIMAR/CIMAR Interdisciplinary Centre of Marine and Environmental Research, U.Porto, 4450-208 Matosinhos, Portugal; (E.S.S.F.); (R.R.); (J.N.F.)
| | - João N. Franco
- CIIMAR/CIMAR Interdisciplinary Centre of Marine and Environmental Research, U.Porto, 4450-208 Matosinhos, Portugal; (E.S.S.F.); (R.R.); (J.N.F.)
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; (Y.H.); (Y.K.)
- Correspondence: (T.N.); (M.M.S.); (L.F.C.C.)
| | - Miguel M. Santos
- CIIMAR/CIMAR Interdisciplinary Centre of Marine and Environmental Research, U.Porto, 4450-208 Matosinhos, Portugal; (E.S.S.F.); (R.R.); (J.N.F.)
- FCUP—Faculty of Sciences, Department of Biology, U.Porto, 4169-007 Porto, Portugal
- Correspondence: (T.N.); (M.M.S.); (L.F.C.C.)
| | - L. Filipe C. Castro
- CIIMAR/CIMAR Interdisciplinary Centre of Marine and Environmental Research, U.Porto, 4450-208 Matosinhos, Portugal; (E.S.S.F.); (R.R.); (J.N.F.)
- FCUP—Faculty of Sciences, Department of Biology, U.Porto, 4169-007 Porto, Portugal
- Correspondence: (T.N.); (M.M.S.); (L.F.C.C.)
| |
Collapse
|
34
|
Cheng V, Dasgupta S, Reddam A, Volz DC. Ciglitazone-a human PPARγ agonist-disrupts dorsoventral patterning in zebrafish. PeerJ 2019; 7:e8054. [PMID: 31741801 PMCID: PMC6858815 DOI: 10.7717/peerj.8054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/17/2019] [Indexed: 01/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-activated transcription factor that regulates lipid/glucose homeostasis and adipocyte differentiation. While the role of PPARγ in adipogenesis and diabetes has been extensively studied, little is known about PPARγ function during early embryonic development. Within zebrafish, maternally-loaded pparγ transcripts are present within the first 6 h post-fertilization (hpf), and de novo transcription of zygotic pparγ commences at ~48 hpf. Since maternal pparγ transcripts are elevated during a critical window of cell fate specification, the objective of this study was to test the hypothesis that PPARγ regulates gastrulation and dorsoventral patterning during zebrafish embryogenesis. To accomplish this objective, we relied on (1) ciglitazone as a potent PPARγ agonist and (2) a splice-blocking, pparγ-specific morpholino to knockdown pparγ. We found that initiation of ciglitazone-a potent human PPARγ agonist-exposure by 4 hpf resulted in concentration-dependent effects on dorsoventral patterning in the absence of epiboly defects during gastrulation, leading to ventralized embryos by 24 hpf. Interestingly, ciglitazone-induced ventralization was reversed by co-exposure with dorsomorphin, a bone morphogenetic protein signaling inhibitor that induces strong dorsalization within zebrafish embryos. Moreover, mRNA-sequencing revealed that lipid- and cholesterol-related processes were affected by exposure to ciglitazone. However, pparγ knockdown did not block ciglitazone-induced ventralization, suggesting that PPARγ is not required for dorsoventral patterning nor involved in ciglitazone-induced toxicity within zebrafish embryos. Our findings point to a novel, PPARγ-independent mechanism of action and phenotype following ciglitazone exposure during early embryonic development.
Collapse
Affiliation(s)
- Vanessa Cheng
- Department of Environmental Sciences, University of California, Riverside, CA, USA
| | - Subham Dasgupta
- Department of Environmental Sciences, University of California, Riverside, CA, USA
| | - Aalekhya Reddam
- Department of Environmental Sciences, University of California, Riverside, CA, USA
| | - David C Volz
- Department of Environmental Sciences, University of California, Riverside, CA, USA
| |
Collapse
|
35
|
Jin M, Zhang B, Sun Y, Zhang S, Li X, Sik A, Bai Y, Zheng X, Liu K. Involvement of peroxisome proliferator-activated receptor γ in anticonvulsant activity of α-asaronol against pentylenetetrazole-induced seizures in zebrafish. Neuropharmacology 2019; 162:107760. [PMID: 31493468 DOI: 10.1016/j.neuropharm.2019.107760] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 07/06/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023]
Abstract
In mammals, peroxisome proliferators activated receptors (PPARs), the nuclear hormone receptors, have been reported to be involved in seizure control. Selective agonists and antagonists of PPARs raise seizure thresholds and suppress seizures, respectively. In this study, we evaluated the anticonvulsant effects of α-asaronol, a metabolic product of α-asarone, on pentylenetetrazole (PTZ)-induced seizures in zebrafish and investigated the underlying mechanisms. As a result, α-asaronol ameliorated seizures with increase of seizure latency, as well as decrease of seizure-like behavior, c-fos expression, and abnormal neuronal discharge in a concentration dependent manner. By comparing gene expression profiles of zebrafish undergoing seizures and α-asaronol pretreated zebrafish, we found that α-asaronol attenuate seizures through increase of PPAR γ expression, while PPAR γ antagonist GW9662 inhibit the anti-seizures actions of α-asaronol. Moreover, molecular docking simulation implied the physical interaction between α-asaronol and PPAR γ. The overall results indicated that the anticonvulsant effects of α-asaronol are regulated through PPAR γ-mediated pathway, which shed light on development of α-asaronol as a potential antiepileptic drug. In addition, it is for first time to report that PPAR γ is associated with seizures in zebrafish, supporting previous evidence that zebrafish is a suitable alternative for studying seizures.
Collapse
Affiliation(s)
- Meng Jin
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789, East Jingshi Road, Ji'nan, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Biosensor of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| | - Baoyue Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789, East Jingshi Road, Ji'nan, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Biosensor of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Ying Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences, Northwest University, Xi'an, 710069, Shanxi Province, PR China; Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi Province, 710069, PR China
| | - Shanshan Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789, East Jingshi Road, Ji'nan, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Biosensor of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China
| | - Xiang Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology of Natural Products (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, NO.44 West Culture Road, Ji'nan, 250012, Shandong Province, PR China
| | - Attila Sik
- Institute of Physiology, Medical School, University of Pecs, Pecs, H-7624, Hungary; Szentagothai Research Centre, University of Pecs, Pecs, H-7624, Hungary; Institute of Clinical Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Yajun Bai
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences, Northwest University, Xi'an, 710069, Shanxi Province, PR China; Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi Province, 710069, PR China.
| | - Xiaohui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, The College of Life Sciences, Northwest University, Xi'an, 710069, Shanxi Province, PR China; Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, Shanxi Province, 710069, PR China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), 28789, East Jingshi Road, Ji'nan, Shandong Province, PR China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China; Key Laboratory for Biosensor of Shandong Province, 28789 East Jingshi Road, Ji'nan, 250103, Shandong Province, PR China.
| |
Collapse
|
36
|
Gomes IDL, Gazo I, Besnardeau L, Hebras C, McDougall A, Dumollard R. Potential roles of nuclear receptors in mediating neurodevelopmental toxicity of known endocrine-disrupting chemicals in ascidian embryos. Mol Reprod Dev 2019; 86:1333-1347. [PMID: 31215734 DOI: 10.1002/mrd.23219] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
Endocrine Disrupting Chemicals (EDCs) are molecules able to interfere with the vertebrate hormonal system in different ways, a major one being the modification of the activity of nuclear receptors (NRs). Several NRs are expressed in the vertebrate brain during embryonic development and these NRs are suspected to be responsible for the neurodevelopmental defects induced by exposure to EDCs in fishes or amphibians and to participate in several neurodevelopmental disorders observed in humans. Known EDCs exert toxicity not only on vertebrate forms of marine life but also on marine invertebrates. However, because hormonal systems of invertebrates are poorly understood, it is not clear whether the teratogenic effects of known EDCs are because of endocrine disruption. The most conserved actors of endocrine systems are the NRs which are present in all metazoan genomes but their functions in invertebrate organisms are still insufficiently characterized. EDCs like bisphenol A have recently been shown to affect neurodevelopment in marine invertebrate chordates called ascidians. Because such phenotypes can be mediated by NRs expressed in the ascidian embryo, we review all the information available about NRs expression during ascidian embryogenesis and discuss their possible involvement in the neurodevelopmental phenotypes induced by EDCs.
Collapse
Affiliation(s)
- Isa D L Gomes
- Sorbonne Université/CNRS, Institut de la Mer, UMR7009 Laboratoire de Biologie du Développement, 06230, Villefranche-sur-Mer, France
| | - Ievgeniia Gazo
- South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, Faculty of Fisheries and Protection of Waters, University of South Bohemia in Ceske Budejovice, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Lydia Besnardeau
- Sorbonne Université/CNRS, Institut de la Mer, UMR7009 Laboratoire de Biologie du Développement, 06230, Villefranche-sur-Mer, France
| | - Céline Hebras
- Sorbonne Université/CNRS, Institut de la Mer, UMR7009 Laboratoire de Biologie du Développement, 06230, Villefranche-sur-Mer, France
| | - Alex McDougall
- Sorbonne Université/CNRS, Institut de la Mer, UMR7009 Laboratoire de Biologie du Développement, 06230, Villefranche-sur-Mer, France
| | - Rémi Dumollard
- Sorbonne Université/CNRS, Institut de la Mer, UMR7009 Laboratoire de Biologie du Développement, 06230, Villefranche-sur-Mer, France
| |
Collapse
|
37
|
Perl E, Waxman JS. Reiterative Mechanisms of Retinoic Acid Signaling during Vertebrate Heart Development. J Dev Biol 2019; 7:jdb7020011. [PMID: 31151214 PMCID: PMC6631158 DOI: 10.3390/jdb7020011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/25/2019] [Accepted: 05/28/2019] [Indexed: 01/07/2023] Open
Abstract
Tightly-regulated levels of retinoic acid (RA) are critical for promoting normal vertebrate development. The extensive history of research on RA has shown that its proper regulation is essential for cardiac progenitor specification and organogenesis. Here, we discuss the roles of RA signaling and its establishment of networks that drive both early and later steps of normal vertebrate heart development. We focus on studies that highlight the drastic effects alternative levels of RA have on early cardiomyocyte (CM) specification and cardiac chamber morphogenesis, consequences of improper RA synthesis and degradation, and known effectors downstream of RA. We conclude with the implications of these findings to our understanding of cardiac regeneration and the etiologies of congenital heart defects.
Collapse
Affiliation(s)
- Eliyahu Perl
- Molecular and Developmental Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
- The Heart Institute and Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Joshua S Waxman
- The Heart Institute and Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
38
|
Baumann L, Segner H, Ros A, Knapen D, Vergauwen L. Thyroid Hormone Disruptors Interfere with Molecular Pathways of Eye Development and Function in Zebrafish. Int J Mol Sci 2019; 20:E1543. [PMID: 30934780 PMCID: PMC6479403 DOI: 10.3390/ijms20071543] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 03/12/2019] [Accepted: 03/25/2019] [Indexed: 02/04/2023] Open
Abstract
The effects of thyroid hormone disrupting chemicals (THDCs) on eye development of zebrafish were investigated. We expected THDC exposure to cause transcriptional changes of vision-related genes, which find their phenotypic anchoring in eye malformations and dysfunction, as observed in our previous studies. Zebrafish were exposed from 0 to 5 days post fertilization (dpf) to either propylthiouracil (PTU), a thyroid hormone synthesis inhibitor, or tetrabromobisphenol-A (TBBPA), which interacts with thyroid hormone receptors. Full genome microarray analyses of RNA isolated from eye tissue revealed that the number of affected transcripts was substantially higher in PTU- than in TBBPA-treated larvae. However, multiple components of phototransduction (e.g., phosphodiesterase, opsins) were responsive to both THDC exposures. Yet, the response pattern for the gene ontology (GO)-class "sensory perception" differed between treatments, with over 90% down-regulation in PTU-exposed fish, compared to over 80% up-regulation in TBBPA-exposed fish. Additionally, the reversibility of effects after recovery in clean water for three days was investigated. Transcriptional patterns in the eyes were still altered and partly overlapped between 5 and 8 dpf, showing that no full recovery occurred within the time period investigated. However, pathways involved in repair mechanisms were significantly upregulated, which indicates activation of regeneration processes.
Collapse
Affiliation(s)
- Lisa Baumann
- Centre for Organismal Studies, Aquatic Ecology and Toxicology, University of Heidelberg, Im Neuenheimer Feld 504, 69120 Heidelberg, Germany.
| | - Helmut Segner
- Vetsuisse Faculty, Centre for Fish and Wildlife Health, University of Bern, Länggassstrasse 122, 3012 Bern, Switzerland.
| | - Albert Ros
- Fischereiforschungsstelle LAZBW, Argenweg 50/1, 88085 Langenargen, Germany.
| | - Dries Knapen
- Department of Veterinary Sciences, Veterinary Physiology and Biochemistry, Zebrafishlab, University of Antwerp, Universiteitsplein 1, 2160 Wilrijk, Belgium.
| | - Lucia Vergauwen
- Department of Veterinary Sciences, Veterinary Physiology and Biochemistry, Zebrafishlab, University of Antwerp, Universiteitsplein 1, 2160 Wilrijk, Belgium.
- Department of Biology, Systemic Physiological and Ecotoxicological Research (SPHERE), University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium.
| |
Collapse
|
39
|
Sant KE, Timme-Laragy AR. Zebrafish as a Model for Toxicological Perturbation of Yolk and Nutrition in the Early Embryo. Curr Environ Health Rep 2019; 5:125-133. [PMID: 29417450 DOI: 10.1007/s40572-018-0183-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Developmental toxicity assessments often focus on structural outcomes and overlook subtle metabolic differences which occur during the early embryonic period. Deviant embryonic nutrition can result in later-life disease, including diabetes, obesity, and cardiovascular disease. Prior to placenta-mediated nutrient exchange, the human embryo requires maternally supplied nutritional substrates for growth, called yolk. Here, we compare the biology of the human and zebrafish yolk and review examples of toxicant-mediated perturbation of yolk defects, composition, and utilization. RECENT FINDINGS Zebrafish embryos, like human embryos, have a protruding yolk sac that serves as a nutritional cache. Aberrant yolk morphology is a common qualitative finding in fish embryotoxicity studies, but quantitative assessment and characterization provides an opportunity to uncover mechanistic targets of toxicant effects on embryonic nutrition. The zebrafish and the study of its yolk sac is an excellent model for uncovering toxicant disruptions to early embryonic nutrition and has potential to discover mechanistic insights into the developmental origins of health and disease.
Collapse
Affiliation(s)
- Karilyn E Sant
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Goessman 171, 686 N Pleasant St, Amherst, MA, 01003, USA
| | - Alicia R Timme-Laragy
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Goessman 171, 686 N Pleasant St, Amherst, MA, 01003, USA.
| |
Collapse
|
40
|
Huo W, Wan R, Wang P, Zhang L, Xia X. Molecular cloning, characterization of dax1 gene and its response to progesterone in Misgurnus anguillicaudatus. Drug Chem Toxicol 2019; 42:624-633. [DOI: 10.1080/01480545.2018.1461900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Weiran Huo
- College of Life Science, Henan Normal University, Xinxiang, Henan, People’s Republic of China
| | - Ruyan Wan
- College of Life Science, Henan Normal University, Xinxiang, Henan, People’s Republic of China
| | - Peijin Wang
- College of Life Science, Henan Normal University, Xinxiang, Henan, People’s Republic of China
| | - Linxia Zhang
- College of Life Science, Henan Normal University, Xinxiang, Henan, People’s Republic of China
| | - Xiaohua Xia
- College of Life Science, Henan Normal University, Xinxiang, Henan, People’s Republic of China
| |
Collapse
|
41
|
Frank D, Sela-Donenfeld D. Hindbrain induction and patterning during early vertebrate development. Cell Mol Life Sci 2019; 76:941-960. [PMID: 30519881 PMCID: PMC11105337 DOI: 10.1007/s00018-018-2974-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 12/28/2022]
Abstract
The hindbrain is a key relay hub of the central nervous system (CNS), linking the bilaterally symmetric half-sides of lower and upper CNS centers via an extensive network of neural pathways. Dedicated neural assemblies within the hindbrain control many physiological processes, including respiration, blood pressure, motor coordination and different sensations. During early development, the hindbrain forms metameric segmented units known as rhombomeres along the antero-posterior (AP) axis of the nervous system. These compartmentalized units are highly conserved during vertebrate evolution and act as the template for adult brainstem structure and function. TALE and HOX homeodomain family transcription factors play a key role in the initial induction of the hindbrain and its specification into rhombomeric cell fate identities along the AP axis. Signaling pathways, such as canonical-Wnt, FGF and retinoic acid, play multiple roles to initially induce the hindbrain and regulate Hox gene-family expression to control rhombomeric identity. Additional transcription factors including Krox20, Kreisler and others act both upstream and downstream to Hox genes, modulating their expression and protein activity. In this review, we will examine the earliest embryonic signaling pathways that induce the hindbrain and subsequent rhombomeric segmentation via Hox and other gene expression. We will examine how these signaling pathways and transcription factors interact to activate downstream targets that organize the segmented AP pattern of the embryonic vertebrate hindbrain.
Collapse
Affiliation(s)
- Dale Frank
- Department of Biochemistry, Faculty of Medicine, The Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, 31096, Haifa, Israel.
| | - Dalit Sela-Donenfeld
- Koret School of Veterinary Medicine, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 76100, Rehovot, Israel.
| |
Collapse
|
42
|
Ortiz-Delgado JB, Funes V, Sarasquete C. The organophosphate pesticide -OP- malathion inducing thyroidal disruptions and failures in the metamorphosis of the Senegalese sole, Solea senegalensis. BMC Vet Res 2019; 15:57. [PMID: 30744622 PMCID: PMC6371575 DOI: 10.1186/s12917-019-1786-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/16/2019] [Indexed: 12/14/2022] Open
Abstract
Background Organophosphate pesticides-OP-, like malathion, can alter the normal functioning of neuro-endocrine systems (e.g., hypothalamus-pituitary-thyroid-HPT- axis), and to interfere on the thyroidal homeostasis. Through direct interactions with thyroid receptors, an/or indirectly via up-stream signalling pathways, from the HPT axis (i.e., negative feedback regulation), malathion possess the ability to affect integrity of thyroidal follicular tissue, and it can also block or delay its hormonal functioning. This insecticide can alter the majority of the ontogenetic processes, inducing several deformities, and also provoking decreases in the growth and survival patterns. The present study has been performed to determine the sublethal effects of malathion during the first month of life of the Senegalese sole, Solea senegalensis, and it is mainly focused on the metamorphosis phase. Different transcript expression levels (i.e. thyroid receptors, matrix and bone -Gla-proteins) and immunohistochemical patterns (i.e. thyroid hormones, osteocalcin, cell proliferation) have been analysed during the most critical phases of the flatfish metamorphosis, that is, through differentiation of thyroid system and skeletal development, migration of the eye, and further adaptation to benthic behaviours. Results In early life stages of the Senegalese sole, the exposure to the highest concentration of malathion (6.25 μg/L) affected to the growth patterns, showing the exposed individuals, a reduction around 60 and 92% of the total length and the dry weigth, respectively. In paralell, a significant reduction of the thyroid follicles (i.e., size and number) it was also been recorded, in a dose-dependent way. Abnormal phenotypes induced in the exposed larvae, did not complete the process of metamorphosis, and displayed several morphological abnormalities and developmental disorders, which were mainly associated with the eye migration process, and with thyroidal and skeletal disorders (i.e., transcriptional and protein changes of thyroid hormones and receptors, and of matrix and bone Gla proteins distribution), that conduced to an inadequate adaptation to the benthic life. Conclusions In the Senegalese sole, the majority of the ontogenetic alterations induced by the exposure to malathion were mainly associated to the metamorphosis period, which is a thyroid-driven proccess. In fact, most crucial and transitional ontogenic events, appeared notably disturbed, for e.g., thyroid gland differentiation and functioning, migration of eye, skeletal development and benthonic behaviors.
Collapse
Affiliation(s)
- Juan B Ortiz-Delgado
- Instituto de Ciencias Marinas de Andalucía-ICMAN, CSIC Campus Universitario Río San Pedro, 11510, Puerto Real, Cádiz, Spain.
| | - Victoria Funes
- IFAPA, Centro el Toruño, Junta de Andalucía, Camino Tiro de Pichón s/n, 11500, El Puerto de Santa María, Cádiz, Spain
| | - Carmen Sarasquete
- Instituto de Ciencias Marinas de Andalucía-ICMAN, CSIC Campus Universitario Río San Pedro, 11510, Puerto Real, Cádiz, Spain
| |
Collapse
|
43
|
Walter KM, Miller GW, Chen X, Yaghoobi B, Puschner B, Lein PJ. Effects of thyroid hormone disruption on the ontogenetic expression of thyroid hormone signaling genes in developing zebrafish (Danio rerio). Gen Comp Endocrinol 2019; 272:20-32. [PMID: 30448381 PMCID: PMC6331280 DOI: 10.1016/j.ygcen.2018.11.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 12/25/2022]
Abstract
Thyroid hormones (THs) regulate neurodevelopment, thus TH disruption is widely posited as a mechanism of developmental neurotoxicity for diverse environmental chemicals. Zebrafish have been proposed as an alternative model for studying the role of TH in developmental neurotoxicity. To realize this goal, it is critical to characterize the normal ontogenetic expression profile of TH signaling molecules in the developing zebrafish and determine the sensitivity of these molecules to perturbations in TH levels. To address these gaps in the existing database, we characterized the transcriptional profiles of TH transporters, deiodinases (DIOs), receptors (TRs), nuclear coactivators (NCOAs), nuclear corepressors (NCORs), and retinoid X receptors (RXRs) in parallel with measurements of endogenous TH concentrations and tshβ mRNA expression throughout the first five days of zebrafish development. Transcripts encoding these TH signaling components were identified and observed to be upregulated around 48-72 h post fertilization (hpf) concurrent with the onset of larval production of T4. Exposure to exogenous T4 and T3 upregulated mct8, dio3-b, trα-a, trβ, and mbp-a levels, and downregulated expression of oatp1c1. Morpholino knockdown of TH transporter mct8 and treatment with 6-propyl-2-thiouracil (PTU) was used to reduce cellular uptake and production of TH, an effect that was associated with downregulation of dio3-b at 120 hpf. Collectively, these data confirm that larval zebrafish express orthologs of TH signaling molecules important in mammalian development and suggest that there may be species differences with respect to impacts of TH disruption on gene transcription.
Collapse
Affiliation(s)
- Kyla M Walter
- Department of Molecular Biosciences, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, United States.
| | - Galen W Miller
- Department of Molecular Biosciences, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, United States.
| | - Xiaopeng Chen
- Department of Molecular Biosciences, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, United States.
| | - Bianca Yaghoobi
- Department of Molecular Biosciences, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, United States.
| | - Birgit Puschner
- Department of Molecular Biosciences, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, United States.
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California-Davis School of Veterinary Medicine, Davis, CA 95616, United States.
| |
Collapse
|
44
|
Lazcano I, Rodríguez-Ortiz R, Villalobos P, Martínez-Torres A, Solís-Saínz JC, Orozco A. Knock-Down of Specific Thyroid Hormone Receptor Isoforms Impairs Body Plan Development in Zebrafish. Front Endocrinol (Lausanne) 2019; 10:156. [PMID: 30930855 PMCID: PMC6427925 DOI: 10.3389/fendo.2019.00156] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 02/21/2019] [Indexed: 12/11/2022] Open
Abstract
The role of thyroid hormones (THs) in development has been extensively studied, however, the specific molecular mechanisms involved are far from being clear. THs act by binding to TH nuclear receptors (TR) that act as ligand-dependent transcription factors to regulate TH-dependent gene expression. Like vertebrates, zebrafish express different isoforms of functional Tr alpha and beta, some of which can bind alternative ligands like 3,5-T2. In this study, we first analyzed the effects of exogenous T3 and 3,5-T2 exposure during embryogenesis. The percentage of affected embryos was similar to those vehicle-injected, suggesting that the early exposure to low TH levels is not sufficient to elicit effects upon the phenotype of the embryo. We then generated crispants for four isoforms of thr to learn more about the role of these receptors in early development. We found that crispant larvae from thraa and a newly identified l-thrb+, but not thrab and canonical thrb1 showed profound deleterious effects upon symmetry and laterality, suggesting early novel roles for these Tr isoforms in the body plan developmental program. Since critical events that determine cell fate start in the late gastrula, we tested if some genes that are expressed during early developmental stages could indeed be TH targets. We identify early development genes, like sox10 and eve, that were specifically over-expressed in thraa and l-thrb+ crispants, suggesting that these specific thr isoforms function as transcription repressors for these genes, while transcription of zic and ets appear to be thraa and l-thrb+-mediated, respectively. Overall, present results show that TH signaling participates in early zebrafish development and identify Tr isoform-specific mediated regulation of early gene expression.
Collapse
Affiliation(s)
- Iván Lazcano
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- Departmento de Investigación Biomédica, Facultad de Medicina, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Roberto Rodríguez-Ortiz
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- CONACYT – Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Mexico
| | - Patricia Villalobos
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Ataúlfo Martínez-Torres
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
| | - Juan Carlos Solís-Saínz
- Departmento de Investigación Biomédica, Facultad de Medicina, Universidad Autónoma de Querétaro, Querétaro, Mexico
| | - Aurea Orozco
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México (UNAM), Querétaro, Mexico
- *Correspondence: Aurea Orozco
| |
Collapse
|
45
|
Li X, Xue Y, Pang L, Len B, Lin Z, Huang J, ShangGuan Z, Pan Y. Agaricus bisporus-derived β-glucan prevents obesity through PPAR γ downregulation and autophagy induction in zebrafish fed by chicken egg yolk. Int J Biol Macromol 2018; 125:820-828. [PMID: 30557642 DOI: 10.1016/j.ijbiomac.2018.12.122] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 01/06/2023]
Abstract
β-(1,4)-d-Glucan with (1,2) and (1,6)-linked branches (short for β-glucan), extracted from Agaricus bisporus (Lange) Sing, had significant anti-obesity and lowering-fat effect. FITC-β-glucan was absorbed by adipocytes of zebrafish larvae when stained by Nile Red. β-Glucan decreased the adiposity mass, reduced the expression of ppar g, mtp, L-fabp, ifabp in ISH, which was coincident as the results of RT-PCT. β-Glucan lowered the level of C/EBP α, c SREBP1, LXR α, PPAR γ by WB analysis, which were accompanied by an increase level in LC3 II/LC3 I and a decline level in p62 in dose-dependent manner. This study explored the effect and mechanisms of Agaricus bisporus derived-β-glucan to regulate lipid metabolism and prevent lipid deposits, and provided the experimental data for its use in diet food and food addictive.
Collapse
Affiliation(s)
- Xiumin Li
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou, Fujian 363000, China.
| | - Yu Xue
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou, Fujian 363000, China
| | - Liang Pang
- The College of Physical Education, Minnan Normal University, Zhangzhou, Fujian 363000, China
| | - Bo Len
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou, Fujian 363000, China
| | - Zhichao Lin
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou, Fujian 363000, China
| | - Jiafu Huang
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou, Fujian 363000, China
| | - Zhaoshui ShangGuan
- Central Laboratory, the First Affiliated Hospital of Xiamen University, Xiamen 361003, China
| | - Yutian Pan
- The Engineering Technological Center of Mushroom Industry, Minnan Normal University, Zhangzhou, Fujian 363000, China.
| |
Collapse
|
46
|
Capitão AMF, Lopes-Marques MS, Ishii Y, Ruivo R, Fonseca ESS, Páscoa I, Jorge RP, Barbosa MAG, Hiromori Y, Miyagi T, Nakanishi T, Santos MM, Castro LFC. Evolutionary Exploitation of Vertebrate Peroxisome Proliferator-Activated Receptor γ by Organotins. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:13951-13959. [PMID: 30398865 DOI: 10.1021/acs.est.8b04399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Globally persistent man-made chemicals display ever-growing ecosystemic consequences, a hallmark of the Anthropocene epoch. In this context, the assessment of how lineage-specific gene repertoires influence organism sensitivity toward endocrine disruptors is a central question in toxicology. A striking example highlights the role of a group of compounds known as obesogens. In mammals, most examples involve the modulation of the nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ). To address the structural and biological determinants of PPARγ exploitation by a model obesogen, tributyltin (TBT), in chordates, we employed comparative genomics, transactivation and ligand binding assays, homology modeling, and site-directed-mutagenesis. We show that the emergence of multiple PPARs (α, β and γ) in vertebrate ancestry coincides with the acquisition of TBT agonist affinity, as can be deduced from the conserved transactivation and binding affinity of the chondrichthyan and mammalian PPARγ. The amphioxus single-copy PPAR is irresponsive to TBT; as well as the investigated teleosts, this is a probable consequence of a specific mutational remodeling of the ligand binding pocket. Our findings endorse the modulatory ability of man-made chemicals and suggest an evolutionarily diverse setting, with impacts for environmental risk assessment.
Collapse
Affiliation(s)
- Ana M F Capitão
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Mónica S Lopes-Marques
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Yoichiro Ishii
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Raquel Ruivo
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Elza S S Fonseca
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Inês Páscoa
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Rodolfo P Jorge
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
| | - Mélanie A G Barbosa
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - Youhei Hiromori
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
- Faculty of Pharmaceutical Sciences , Suzuka University of Medical Science 3500-3 Minamitamagaki , Suzuka , Mie 513-8670 , Japan
| | - Takayuki Miyagi
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology , Gifu Pharmaceutical University , 1-25-4 Daigaku-nishi , Gifu , Gifu 501-1196 , Japan
| | - Miguel M Santos
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| | - L Filipe C Castro
- Interdisciplinary Centre of Marine and Environmental Research , University of Porto , 4450-208 Matosinhos , Portugal
- Department of Biology, Faculty of Sciences , University of Porto , 4169-007 Porto , Portugal
| |
Collapse
|
47
|
Hsieh YC, Chiang MC, Huang YC, Yeh TH, Shih HY, Liu HF, Chen HY, Wang CP, Cheng YC. Pparα deficiency inhibits the proliferation of neuronal and glial precursors in the zebrafish central nervous system. Dev Dyn 2018; 247:1264-1275. [DOI: 10.1002/dvdy.24683] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/12/2018] [Accepted: 10/16/2018] [Indexed: 12/28/2022] Open
Affiliation(s)
- Yen-Che Hsieh
- Graduate Institute of Biomedical Sciences, College of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Ming-Chang Chiang
- Department of Life Science; Fu Jen Catholic University; New Taipei City Taiwan
| | - Yin-Cheng Huang
- College of Medicine; Chang Gung University; Taoyuan Taiwan
- Department of Neurosurgery; Chang Gung Memorial Hospital; Linkou, Taoyuan Taiwan
| | - Tu-Hsueh Yeh
- College of Medicine; Chang Gung University; Taoyuan Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital; Linkou, Taoyuan Taiwan
- Section of Movement Disorders, Department of Neurology; Chang Gung Memorial Hospital; Linkou, Taoyuan Taiwan
- Department of Neurology; Taipei Medical University Hospital; Taipei Taiwan
| | - Hung-Yu Shih
- Graduate Institute of Biomedical Sciences, College of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Han-Fang Liu
- Graduate Institute of Biomedical Sciences, College of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Hao-Yuan Chen
- Graduate Institute of Biomedical Sciences, College of Medicine; Chang Gung University; Taoyuan Taiwan
| | - Chien-Ping Wang
- School of Medicine, College of Medicine, Chang Gung University; Taoyuan Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine; Chang Gung University; Taoyuan Taiwan
- College of Medicine; Chang Gung University; Taoyuan Taiwan
- Neuroscience Research Center, Chang Gung Memorial Hospital; Linkou, Taoyuan Taiwan
| |
Collapse
|
48
|
Chambers JM, Poureetezadi SJ, Addiego A, Lahne M, Wingert RA. ppargc1a controls nephron segmentation during zebrafish embryonic kidney ontogeny. eLife 2018; 7:40266. [PMID: 30475208 PMCID: PMC6279350 DOI: 10.7554/elife.40266] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/23/2018] [Indexed: 02/06/2023] Open
Abstract
Nephron segmentation involves a concert of genetic and molecular signals that are not fully understood. Through a chemical screen, we discovered that alteration of peroxisome proliferator-activated receptor (PPAR) signaling disrupts nephron segmentation in the zebrafish embryonic kidney (Poureetezadi et al., 2016). Here, we show that the PPAR co-activator ppargc1a directs renal progenitor fate. ppargc1a mutants form a small distal late (DL) segment and an expanded proximal straight tubule (PST) segment. ppargc1a promotes DL fate by regulating the transcription factor tbx2b, and restricts expression of the transcription factor sim1a to inhibit PST fate. Interestingly, sim1a restricts ppargc1a expression to promote the PST, and PST development is fully restored in ppargc1a/sim1a-deficient embryos, suggesting Ppargc1a and Sim1a counterbalance each other in an antagonistic fashion to delineate the PST segment boundary during nephrogenesis. Taken together, our data reveal new roles for Ppargc1a during development, which have implications for understanding renal birth defects.
Collapse
Affiliation(s)
- Joseph M Chambers
- Department of Biological Sciences, University of Notre Dame, Indiana, United States.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Indiana, United States.,Center for Zebrafish Research, University of Notre Dame, Indiana, United States
| | - Shahram Jevin Poureetezadi
- Department of Biological Sciences, University of Notre Dame, Indiana, United States.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Indiana, United States.,Center for Zebrafish Research, University of Notre Dame, Indiana, United States
| | - Amanda Addiego
- Department of Biological Sciences, University of Notre Dame, Indiana, United States.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Indiana, United States.,Center for Zebrafish Research, University of Notre Dame, Indiana, United States
| | - Manuela Lahne
- Department of Biological Sciences, University of Notre Dame, Indiana, United States.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Indiana, United States.,Center for Zebrafish Research, University of Notre Dame, Indiana, United States
| | - Rebecca A Wingert
- Department of Biological Sciences, University of Notre Dame, Indiana, United States.,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Indiana, United States.,Center for Zebrafish Research, University of Notre Dame, Indiana, United States
| |
Collapse
|
49
|
Xia X, Huo W, Wan R, Wang P, Chang Z. Cloning, characterization and function analysis of DAX1 in Chinese loach (Paramisgurnus dabryanus). Genetica 2018; 146:487-496. [PMID: 30206752 DOI: 10.1007/s10709-018-0039-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 09/06/2018] [Indexed: 02/02/2023]
Abstract
The mechanisms of sex determination and differentiation have not been elucidated in most fish species. In this study, the full-length cDNAs of DAX1 was cloned and characterized in aquaculture fish Chinese loach (Paramisgurnus dabryanus), designated as Pd-DAX1. The cDNA sequence of Pd-DAX1 was 1261 bp, including 795 bp open reading frame (ORF) encoding 264 amino acids. Pd-DAX1 shares highly identical sequence with DAX1 homologues from different species. The expression profiles of Pd-DAX1 in different developmental stages and diverse adult tissues were analyzed by quantitative real-time RT-PCR and in situ hybridization (ISH). Pd-DAX1 was continuously expressed during embryogenesis, with the extensive distribution in the development of the central nervous system. Tissue distribution analysis revealed that Pd-DAX1 expressed widely in adult tissues, with the highest expression level found in testis, moderate level in ovary, showing a sex-dimorphic expression pattern. Pd-DAX1 mainly located in spermatogonia cells, spermatocytes, primary oocytes and previtellogenic oocyte cells, implying that Pd-DAX1 may involve in gametogenesis. These preliminary findings suggest that Pd-DAX1 gene is highly conserved during vertebrate evolution and involved in a wide range of developmental processes including embryogenesis, central nervous system development and gonad development.
Collapse
Affiliation(s)
- Xiaohua Xia
- Molecular and Genetic Laboratory, College of Life Science, Henan Normal University, 46# East of Construction Road, Xinxiang, 453007, Henan, People's Republic of China.
| | - Weiran Huo
- Molecular and Genetic Laboratory, College of Life Science, Henan Normal University, 46# East of Construction Road, Xinxiang, 453007, Henan, People's Republic of China
| | - Ruyan Wan
- Molecular and Genetic Laboratory, College of Life Science, Henan Normal University, 46# East of Construction Road, Xinxiang, 453007, Henan, People's Republic of China
| | - Peijin Wang
- Molecular and Genetic Laboratory, College of Life Science, Henan Normal University, 46# East of Construction Road, Xinxiang, 453007, Henan, People's Republic of China
| | - Zhongjie Chang
- Molecular and Genetic Laboratory, College of Life Science, Henan Normal University, 46# East of Construction Road, Xinxiang, 453007, Henan, People's Republic of China
| |
Collapse
|
50
|
Independent losses of a xenobiotic receptor across teleost evolution. Sci Rep 2018; 8:10404. [PMID: 29991818 PMCID: PMC6039460 DOI: 10.1038/s41598-018-28498-4] [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: 02/16/2018] [Accepted: 06/22/2018] [Indexed: 01/25/2023] Open
Abstract
Sensitivity to environmental stressors largely depend on the genetic complement of the organism. Recent sequencing and assembly of teleost fish genomes enable us to trace the evolution of defense genes in the largest and most diverse group of vertebrates. Through genomic searches and in-depth analysis of gene loci in 76 teleost genomes, we show here that the xenosensor pregnane X receptor (Pxr, Nr1i2) is absent in more than half of these species. Notably, out of the 27 genome assemblies that belong to the Gadiformes order, the pxr gene was only retained in the Merluccidae family (hakes) and Pelagic cod (Melanonus zugmayeri). As an important receptor for a wide range of drugs and environmental pollutants, vertebrate PXR regulate the transcription of a number of genes involved in the biotransformation of xenobiotics, including cytochrome P450 enzymes (CYP). In the absence of Pxr, we suggest that the aryl hydrocarbon receptor (Ahr) have evolved an extended regulatory role by governing the expression of certain Pxr target genes, such as cyp3a, in Atlantic cod (Gadus morhua). However, as several independent losses of pxr have occurred during teleost evolution, other lineages and species may have adapted alternative compensating mechanisms for controlling crucial cellular defense mechanisms.
Collapse
|