1
|
Lin X, Zhao Y, Bai Y, Meng K, Chen Y, Hu M, Liu F, Luo D. Integrating gonadal RNA-seq and small RNA-seq to analyze mRNA and miRNA changes in medaka sex differentiation. Sci Data 2025; 12:778. [PMID: 40355444 PMCID: PMC12069655 DOI: 10.1038/s41597-025-05129-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025] Open
Abstract
MicroRNAs are important post-transcriptional regulators, yet the molecular crosstalk between miRNAs and their target genes during sex differentiation remains poorly understood. Medaka (Oryzias latipes), the first fish in which the sex determination gene was identified, serves as an ideal model for studying this process. Here, we generated gonadal RNA-seq and small RNA-seq data from XYDMY- females, wild-type females and males to explore this crosstalk. A total of twenty-seven RNA-seq datasets, comprising 188 Gb of raw reads, and twenty-seven small RNA-seq datasets, totaling 18 Gb of raw reads, were collected, covering 10, 30 and 120 days. After optimizing the mapping and normalizing, we conducted transcriptional and post-transcriptional dynamic analyses of differentially expressed genes and miRNAs between WT females and males, as well as between WT females and XYDMY- females. Additionally, we integrated the RNA-seq and small RNA-seq data to construct comprehensive interaction networks and performed a detailed analysis of the temporal dynamics in gene and miRNA expression. These resources offer valuable insights into the transcriptional regulation of gonadal differentiation and development in vertebrates.
Collapse
Affiliation(s)
- Xing Lin
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yuanli Zhao
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Yifan Bai
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaifeng Meng
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yuanyuan Chen
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, 430072, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Meidi Hu
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, 430072, China
- Fisheries College, Ocean University of China, Qingdao, 266001, China
| | - Fei Liu
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Daji Luo
- State Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture, Institute of Hydrobiology, Hubei Hongshan Laboratory, Chinese Academy of Sciences, Wuhan, 430072, China.
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
- Fisheries College, Ocean University of China, Qingdao, 266001, China.
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
2
|
Zhang Y, Wang J, Fang H, Hu S, Yang B, Zhou J, Grifone R, Li P, Lu T, Wang Z, Zhang C, Huang Y, Wu D, Gong Q, Shi DL, Li A, Shao M. Rbm24a dictates mRNA recruitment for germ granule assembly in zebrafish. EMBO J 2025:10.1038/s44318-025-00442-z. [PMID: 40281355 DOI: 10.1038/s44318-025-00442-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 03/15/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025] Open
Abstract
The germ granules are ribonucleoprotein (RNP) biomolecular condensates that determine the fate of primordial germ cells (PGCs) and serve as a model for studying RNP granule assembly. Here, we show that the maternal RNA-binding protein Rbm24a is a key factor governing the specific sorting of mRNAs into germ granules. Mechanistically, Rbm24a interacts with the germ plasm component Buc to dictate the specific recruitment of germ plasm mRNAs into phase-separated condensates. Germ plasm particles lacking Rbm24a and mRNAs fail to undergo kinesin-dependent transport toward cleavage furrows where small granules fuse into large aggregates. Therefore, the loss of maternal Rbm24a causes a complete degradation of the germ plasm and the disappearance of PGCs. These findings demonstrate that the Rbm24a/Buc complex functions as a nucleating organizer of germ granules, highlighting an emerging mechanism for RNA-binding proteins in reading and recruiting RNA components into a phase-separated protein scaffold.
Collapse
Affiliation(s)
- Yizhuang Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, 266237, Qingdao, China
| | - Jiasheng Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, 266237, Qingdao, China
| | - Hailing Fang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, 266237, Qingdao, China
| | - Shuqi Hu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
| | - Boya Yang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, 266237, Qingdao, China
| | - Jiayi Zhou
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, 266237, Qingdao, China
| | - Raphaëlle Grifone
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Paris, France
| | - Panfeng Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
| | - Tong Lu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, 266237, Qingdao, China
| | - Zhengyang Wang
- Shandong University Taishan College, 266237, Qingdao, China
| | - Chong Zhang
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, 524045, Zhanjiang, China
| | - Yubin Huang
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, 266237, Qingdao, China
| | - Dalei Wu
- State Key Laboratory of Microbial Technology, Institute of Microbial Technology, 266237, Qingdao, China
| | - Qianqian Gong
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, 266237, Qingdao, China
| | - De-Li Shi
- Sorbonne Université, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8263, INSERM U1345, Development, Adaptation and Ageing, Paris, France.
- Fang Zongxi Center, Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China.
| | - Ang Li
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China.
| | - Ming Shao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences and Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 266237, Qingdao, China.
- Key Laboratory for Experimental Teratology of the Ministry of Education, Shandong University, 266237, Qingdao, China.
- Shandong University-Yuanchen Joint Biomedical Technology Laboratory, 266237, Qingdao, China.
| |
Collapse
|
3
|
Kojima ML, Hoppe C, Giraldez AJ. The maternal-to-zygotic transition: reprogramming of the cytoplasm and nucleus. Nat Rev Genet 2025; 26:245-267. [PMID: 39587307 PMCID: PMC11928286 DOI: 10.1038/s41576-024-00792-0] [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] [Accepted: 10/08/2024] [Indexed: 11/27/2024]
Abstract
A fertilized egg is initially transcriptionally silent and relies on maternally provided factors to initiate development. For embryonic development to proceed, the oocyte-inherited cytoplasm and the nuclear chromatin need to be reprogrammed to create a permissive environment for zygotic genome activation (ZGA). During this maternal-to-zygotic transition (MZT), which is conserved in metazoans, transient totipotency is induced and zygotic transcription is initiated to form the blueprint for future development. Recent technological advances have enhanced our understanding of MZT regulation, revealing common themes across species and leading to new fundamental insights about transcription, mRNA decay and translation.
Collapse
Affiliation(s)
- Mina L Kojima
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Caroline Hoppe
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Antonio J Giraldez
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Wang Z, Wang S, Bi Y, Boiti A, Zhang S, Vallone D, Lan X, Foulkes NS, Zhao H. Light-regulated microRNAs shape dynamic gene expression in the zebrafish circadian clock. PLoS Genet 2025; 21:e1011545. [PMID: 39777894 PMCID: PMC11750094 DOI: 10.1371/journal.pgen.1011545] [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: 06/20/2024] [Revised: 01/21/2025] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
A key property of the circadian clock is that it is reset by light to remain synchronized with the day-night cycle. An attractive model to explore light input to the circadian clock in vertebrates is the zebrafish. Circadian clocks in zebrafish peripheral tissues and even zebrafish-derived cell lines are entrainable by direct light exposure thus providing unique insight into the function and evolution of light regulatory pathways. Our previous work has revealed that light-induced gene transcription is a key step in the entrainment of the circadian clock as well as enabling the more general adaptation of zebrafish cells to sunlight exposure. However, considerable evidence points to post-transcriptional regulatory mechanisms, notably microRNAs (miRNAs), playing an essential role in shaping dynamic changes in mRNA levels. Therefore, does light directly impact the function of miRNAs? Are there light-regulated miRNAs and if so, which classes of mRNA do they target? To address these questions, we performed a complete sequencing analysis of light-induced changes in the zebrafish transcriptome, encompassing small non-coding RNAs as well as mRNAs. Importantly, we identified sets of light-regulated miRNAs, with many regulatory targets representing light-inducible mRNAs including circadian clock genes and genes involved in redox homeostasis. We subsequently focused on the light-responsive miR-204-3-3p and miR-430a-3p which are predicted to regulate the expression of cryptochrome genes (cry1a and cry1b). Luciferase reporter assays validated the target binding of miR-204-3-3p and miR-430a-3p to the 3'UTRs of cry1a and cry1b, respectively. Furthermore, treatment with mimics and inhibitors of these two miRNAs significantly affected the dynamic expression of their target genes but also other core clock components (clock1a, bmal1b, per1b, per2, per3), as well as the rhythmic locomotor activity of zebrafish larvae. Thus, our identification of light-responsive miRNAs reveals new intricacy in the multi-level regulation of the circadian clockwork by light.
Collapse
Affiliation(s)
- Zuo Wang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| | - Shuang Wang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| | - Yi Bi
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Alessandra Boiti
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Shengxiang Zhang
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| | - Daniela Vallone
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Xianyong Lan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Nicholas S. Foulkes
- Institute of Biological and Chemical Systems, Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT), Eggenstein-Leopoldshafen, Germany
| | - Haiyu Zhao
- School of Life Sciences, Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou, China
| |
Collapse
|
5
|
Rosero J, Monzani PS, Pessoa GP, Coelho GCZ, Carvalho GB, López LS, Senhorini JA, Dos Santos SCA, Yasui GS. Traceability of primordial germ cells in three neotropical fish species aiming genetic conservation actions. FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:2025-2042. [PMID: 38060079 DOI: 10.1007/s10695-023-01279-1] [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: 07/13/2023] [Accepted: 11/24/2023] [Indexed: 12/08/2023]
Abstract
Primordial germ cells (PGCs) are embryonic pluripotent cells that can differentiate into spermatogonia and oogonia, and therefore, PGCs are a genetic source for germplasm conservation through cryobanking and the generation of germline chimeras. The knowledge of PGC migration routes is essential for transplantation studies. In this work, the mRNA synthesized from the ddx4 3'UTR sequence of Pseudopimelodus mangurus, in fusion with gfp or dsred, was microinjected into zygotes of three neotropical species (P. mangurus, Astyanax altiparanae, and Prochilodus lineatus) for PGC labeling. Visualization of labeled PGCs was achieved by fluorescence microscopy during embryonic development. In addition, ddx4 and dnd1 expressions were evaluated during embryonic development, larvae, and adult tissues of P. mangurus, to validate their use as a PGC marker. As a result, the effective identification of presumptive PGCs was obtained. DsRed-positive PGC of P. mangurus was observed in the hatching stage, GFP-positive PGC of A. altiparanae in the gastrula stage, and GFP-positive PGCs from P. lineatus were identified at the segmentation stage, with representative labeling percentages of 29% and 16% in A. altiparanae and P. lineatus, respectively. The expression of ddx4 and dnd1 of P. mangurus confirmed the specificity of these genes in germ cells. These results point to the functionality of the P. mangurus ddx4 3'UTR sequence as a PGC marker, demonstrating that PGC labeling was more efficient in A. altiparanae and P. lineatus. The procedures used to identify PGCs in P. mangurus consolidate the first step for generating germinal chimeras as a conservation action of P. mangurus.
Collapse
Affiliation(s)
- Jenyffer Rosero
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil.
- Laboratory of Fish Biotechnology, National Center for Research and Conservation of Continental Aquatic Biodiversity, Chico Mendes Institute of Biodiversity Conservation, Pirassununga, São Paulo, Brazil.
| | - Paulo Sérgio Monzani
- Laboratory of Fish Biotechnology, National Center for Research and Conservation of Continental Aquatic Biodiversity, Chico Mendes Institute of Biodiversity Conservation, Pirassununga, São Paulo, Brazil
- Institute of Bioscience, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Giselle Pessanha Pessoa
- Laboratory of Fish Biotechnology, National Center for Research and Conservation of Continental Aquatic Biodiversity, Chico Mendes Institute of Biodiversity Conservation, Pirassununga, São Paulo, Brazil
- Institute of Bioscience, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Geovanna Carla Zacheo Coelho
- Laboratory of Fish Biotechnology, National Center for Research and Conservation of Continental Aquatic Biodiversity, Chico Mendes Institute of Biodiversity Conservation, Pirassununga, São Paulo, Brazil
- Institute of Bioscience, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Gabriella Braga Carvalho
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil
- Laboratory of Fish Biotechnology, National Center for Research and Conservation of Continental Aquatic Biodiversity, Chico Mendes Institute of Biodiversity Conservation, Pirassununga, São Paulo, Brazil
| | - Lucia Suárez López
- Laboratory of Fish Biotechnology, National Center for Research and Conservation of Continental Aquatic Biodiversity, Chico Mendes Institute of Biodiversity Conservation, Pirassununga, São Paulo, Brazil
- Institute of Bioscience, São Paulo State University, Botucatu, São Paulo, Brazil
| | - José Augusto Senhorini
- Laboratory of Fish Biotechnology, National Center for Research and Conservation of Continental Aquatic Biodiversity, Chico Mendes Institute of Biodiversity Conservation, Pirassununga, São Paulo, Brazil
- Institute of Bioscience, São Paulo State University, Botucatu, São Paulo, Brazil
| | | | - George Shigueki Yasui
- Department of Animal Reproduction, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, São Paulo, Brazil
- Laboratory of Fish Biotechnology, National Center for Research and Conservation of Continental Aquatic Biodiversity, Chico Mendes Institute of Biodiversity Conservation, Pirassununga, São Paulo, Brazil
| |
Collapse
|
6
|
Shi DL. Interplay of RNA-binding proteins controls germ cell development in zebrafish. J Genet Genomics 2024; 51:889-899. [PMID: 38969260 DOI: 10.1016/j.jgg.2024.06.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/07/2024]
Abstract
The specification of germ cells in zebrafish mostly relies on an inherited mechanism by which localized maternal determinants, called germ plasm, confer germline fate in the early embryo. Extensive studies have partially allowed the identification of key regulators governing germ plasm formation and subsequent germ cell development. RNA-binding proteins, acting in concert with other germ plasm components, play essential roles in the organization of the germ plasm and the specification, migration, maintenance, and differentiation of primordial germ cells. The loss of their functions impairs germ cell formation and causes sterility or sexual conversion. Evidence is emerging that they instruct germline development through differential regulation of mRNA fates in somatic and germ cells. However, the challenge remains to decipher the complex interplay of maternal germ plasm components in germ plasm compartmentalization and germ cell specification. Because failure to control the developmental outcome of germ cells disrupts the formation of gametes, it is important to gain a complete picture of regulatory mechanisms operating in the germ cell lineage. This review sheds light on the contributions of RNA-binding proteins to germ cell development in zebrafish and highlights intriguing questions that remain open for future investigation.
Collapse
Affiliation(s)
- De-Li Shi
- Laboratory of Developmental Biology, CNRS-UMR7622, Institut de Biologie Paris-Seine (IBPS), Sorbonne University, 75005 Paris, France.
| |
Collapse
|
7
|
Fishman L, Modak A, Nechooshtan G, Razin T, Erhard F, Regev A, Farrell JA, Rabani M. Cell-type-specific mRNA transcription and degradation kinetics in zebrafish embryogenesis from metabolically labeled single-cell RNA-seq. Nat Commun 2024; 15:3104. [PMID: 38600066 PMCID: PMC11006943 DOI: 10.1038/s41467-024-47290-9] [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: 04/12/2023] [Accepted: 03/27/2024] [Indexed: 04/12/2024] Open
Abstract
During embryonic development, pluripotent cells assume specialized identities by adopting particular gene expression profiles. However, systematically dissecting the relative contributions of mRNA transcription and degradation to shaping those profiles remains challenging, especially within embryos with diverse cellular identities. Here, we combine single-cell RNA-Seq and metabolic labeling to capture temporal cellular transcriptomes of zebrafish embryos where newly-transcribed (zygotic) and pre-existing (maternal) mRNA can be distinguished. We introduce kinetic models to quantify mRNA transcription and degradation rates within individual cell types during their specification. These models reveal highly varied regulatory rates across thousands of genes, coordinated transcription and destruction rates for many transcripts, and link differences in degradation to specific sequence elements. They also identify cell-type-specific differences in degradation, namely selective retention of maternal transcripts within primordial germ cells and enveloping layer cells, two of the earliest specified cell types. Our study provides a quantitative approach to study mRNA regulation during a dynamic spatio-temporal response.
Collapse
Affiliation(s)
- Lior Fishman
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9190401, Israel
| | - Avani Modak
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, 20814, USA
| | - Gal Nechooshtan
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9190401, Israel
| | - Talya Razin
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9190401, Israel
| | - Florian Erhard
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
- Chair of Computational Immunology, University of Regensburg, Regensburg, Germany
| | - Aviv Regev
- Department of Biology, MIT, Cambridge, MA, 02139, USA
- Klarman Cell Observatory Broad Institute of MIT and Harvard Cambridge, Cambridge, MA, 02142, USA
| | - Jeffrey A Farrell
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, 20814, USA.
| | - Michal Rabani
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9190401, Israel.
| |
Collapse
|
8
|
Fishman L, Nechooshtan G, Erhard F, Regev A, Farrell JA, Rabani M. Single-cell temporal dynamics reveals the relative contributions of transcription and degradation to cell-type specific gene expression in zebrafish embryos. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.537620. [PMID: 37131717 PMCID: PMC10153228 DOI: 10.1101/2023.04.20.537620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
During embryonic development, pluripotent cells assume specialized identities by adopting particular gene expression profiles. However, systematically dissecting the underlying regulation of mRNA transcription and degradation remains a challenge, especially within whole embryos with diverse cellular identities. Here, we collect temporal cellular transcriptomes of zebrafish embryos, and decompose them into their newly-transcribed (zygotic) and pre-existing (maternal) mRNA components by combining single-cell RNA-Seq and metabolic labeling. We introduce kinetic models capable of quantifying regulatory rates of mRNA transcription and degradation within individual cell types during their specification. These reveal different regulatory rates between thousands of genes, and sometimes between cell types, that shape spatio-temporal expression patterns. Transcription drives most cell-type restricted gene expression. However, selective retention of maternal transcripts helps to define the gene expression profiles of germ cells and enveloping layer cells, two of the earliest specified cell-types. Coordination between transcription and degradation restricts expression of maternal-zygotic genes to specific cell types or times, and allows the emergence of spatio-temporal patterns when overall mRNA levels are held relatively constant. Sequence-based analysis links differences in degradation to specific sequence motifs. Our study reveals mRNA transcription and degradation events that control embryonic gene expression, and provides a quantitative approach to study mRNA regulation during a dynamic spatio-temporal response.
Collapse
Affiliation(s)
- Lior Fishman
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9190401, Israel
| | - Gal Nechooshtan
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9190401, Israel
| | - Florian Erhard
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Aviv Regev
- Department of Biology, MIT, Cambridge MA 02139, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Jeffrey A. Farrell
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, 20814, USA
| | - Michal Rabani
- Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Jerusalem, 9190401, Israel
| |
Collapse
|
9
|
Li W, Liu W, Mo C, Yi M, Gui J. Two Novel lncRNAs Regulate Primordial Germ Cell Development in Zebrafish. Cells 2023; 12:cells12040672. [PMID: 36831339 PMCID: PMC9954370 DOI: 10.3390/cells12040672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 02/22/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) are regulatory transcripts in various biological processes. However, the role of lncRNAs in germline development remains poorly understood, especially for fish primordial germ cell (PGC) development. In this study, the lncRNA profile of zebrafish PGC was revealed by single cell RNA-sequencing and bioinformatic prediction. We established the regulation network of lncRNA-mRNA associated with PGC development, from which we identified three novel lncRNAs-lnc172, lnc196, and lnc304-highly expressing in PGCs and gonads. Fluorescent in situ hybridization indicated germline-specific localization of lnc196 and lnc304 in the cytoplasm and nucleus of spermatogonia, spermatocyte, and occyte, and they were co-localized with vasa in the cytoplasm of the spermatogonia. By contrast, lnc172 was localized in the cytoplasm of male germline, myoid cells and ovarian somatic cells. Loss- and gain-of-function experiments demonstrated that knockdown and PGC-specific overexpression of lnc304 as well as universal overexpression of lnc172 significantly disrupted PGC development. In summary, the present study revealed the lncRNA profile of zebrafish PGC and identified two novel lncRNAs associated with PGC development, providing new insights for understanding the regulatory mechanism of PGC development.
Collapse
Affiliation(s)
- Wenjing Li
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
| | - Wei Liu
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
| | - Chengyu Mo
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
| | - Meisheng Yi
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
- Correspondence: (M.Y.); (J.G.)
| | - Jianfang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 420072, China
- Correspondence: (M.Y.); (J.G.)
| |
Collapse
|
10
|
Shiguemoto GF, Coelho GCZ, López LS, Pessoa GP, Dos Santos SCA, Senhorini JA, Monzani PS, Yasui GS. Primordial germ cell identification and traceability during the initial development of the Siluriformes fish Pseudopimelodus mangurus. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:1137-1153. [PMID: 35925505 DOI: 10.1007/s10695-022-01106-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/23/2022] [Indexed: 06/15/2023]
Abstract
Primordial germ cells (PGCs) are responsible for generating all germ cells. Therefore, they are essential targets to be used as a tool for the production of germline chimeras. The labeling and route of PGCs were evaluated during the initial embryonic development of Pseudopimelodus mangurus, using whole-mount in situ hybridization (WISH) and mRNA microinjection in zygotes. A specific antisense RNA probe constituted by a partial coding region from P. mangurus nanos3 mRNA was synthesized for the WISH method. RNA microinjection was performed using the GFP gene reporter regulated by translation regulatory P. mangurus buc and nanos3 3'UTR sequences, germline-specific markers used to describe in vivo migration of PGCs. Nanos3 and buc gene expression was evaluated in tissues for male and female adults and initial development phases and larvae from the first to seventh days post-hatching. The results from the WISH technique indicated the origin of PGCs in P. mangurus from the aggregations of nanos3 mRNA in the cleavage grooves and the signals obtained from nanos3 probes corresponded topographically to the migratory patterns of the PGCs reported for other fish species. Diffuse signals were observed in all blastomeres until the 16-cell stage, which could be related to the two sequences of the nanos3 3'UTR observed in the P. mangurus unfertilized egg transcriptome. Microinjection was not successful using GFP-Dr-nanos1 3'UTR mRNA and GFP-Pm-buc 3'UTR mRNA and allowed the identification of potential PGCs with less than 2% efficiency only and after hatching using GFP-Pm-nanos3 3'UTR. Nanos3 and buc gene expression was reported in the female gonads and from fertilized eggs until the blastula phase. These results provide information about the PGC migration of P. mangurus and the possible use of PGCs for the future generation of germline chimeras to be applied in the conservation efforts of Neotropical Siluriformes species. This study can contribute to establishing genetic banks, manipulating organisms, and assisting in biotechnologies such as transplanting germ cells in fish.
Collapse
Affiliation(s)
- Gustavo Fonseca Shiguemoto
- Institute of Bioscience, São Paulo State University, Botucatu, SP, Brazil
- Laboratory of Fish Biotechnology, Chico Mendes Institute for Biodiversity Conservation /National Center for Research and Conservation of Continental Aquatic Biodiversity, Pirassununga, SP, Brazil
| | - Geovanna Carla Zacheo Coelho
- Institute of Bioscience, São Paulo State University, Botucatu, SP, Brazil
- Laboratory of Fish Biotechnology, Chico Mendes Institute for Biodiversity Conservation /National Center for Research and Conservation of Continental Aquatic Biodiversity, Pirassununga, SP, Brazil
| | - Lucia Suárez López
- Institute of Bioscience, São Paulo State University, Botucatu, SP, Brazil
- Laboratory of Fish Biotechnology, Chico Mendes Institute for Biodiversity Conservation /National Center for Research and Conservation of Continental Aquatic Biodiversity, Pirassununga, SP, Brazil
| | - Giselle Pessanha Pessoa
- Institute of Bioscience, São Paulo State University, Botucatu, SP, Brazil
- Laboratory of Fish Biotechnology, Chico Mendes Institute for Biodiversity Conservation /National Center for Research and Conservation of Continental Aquatic Biodiversity, Pirassununga, SP, Brazil
| | | | - José Augusto Senhorini
- Institute of Bioscience, São Paulo State University, Botucatu, SP, Brazil
- Laboratory of Fish Biotechnology, Chico Mendes Institute for Biodiversity Conservation /National Center for Research and Conservation of Continental Aquatic Biodiversity, Pirassununga, SP, Brazil
| | - Paulo Sérgio Monzani
- Institute of Bioscience, São Paulo State University, Botucatu, SP, Brazil.
- Laboratory of Fish Biotechnology, Chico Mendes Institute for Biodiversity Conservation /National Center for Research and Conservation of Continental Aquatic Biodiversity, Pirassununga, SP, Brazil.
| | - George Shigueki Yasui
- Institute of Bioscience, São Paulo State University, Botucatu, SP, Brazil
- Laboratory of Fish Biotechnology, Chico Mendes Institute for Biodiversity Conservation /National Center for Research and Conservation of Continental Aquatic Biodiversity, Pirassununga, SP, Brazil
| |
Collapse
|
11
|
Song W, Gan W, Xie Z, Chen J, Wang L. Small RNA sequencing reveals sex-related miRNAs in Collichthys lucidus. Front Genet 2022; 13:955645. [PMID: 36092867 PMCID: PMC9458855 DOI: 10.3389/fgene.2022.955645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
Collichthys lucidus (C. lucidus) is an economically important fish species, exhibiting sexual dimorphism in its growth rate. However, there is a lack of research on its underlying sex-related mechanisms. Therefore, small RNA sequencing was performed to better comprehend these sex-related molecular mechanisms. In total, 171 differentially expressed miRNAs (DE-miRNAs) were identified between the ovaries and testes. Functional enrichment analysis revealed that the target genes of DE-miRNAs were considerably enriched in the p53 signaling, PI3K–Akt signaling, and TGF-beta signaling pathways. In addition, sex-related miRNAs were identified, and the expression of miR-430c-3p and miR-430f-3p was specifically observed in the gonads compared with other organs and their expression was markedly upregulated in the testes relative to the ovaries. Bmp15 was a target of miR-430c-3p and was greatly expressed in the ovaries compared with the testes. Importantly, miR-430c-3p and bmp15 co-expressed in the ovaries and testes. This research provides the first detailed miRNA profiles for C. lucidus concerning sex, likely laying the basis for further studies on sex differentiation in C. lucidus.
Collapse
Affiliation(s)
- Wei Song
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Wu Gan
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, China
| | - Zhengli Xie
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
| | - Jia Chen
- State Key Laboratory of Large Yellow Croaker Breeding, Fuding Seagull Fishing Food Co. Ltd., Ningde, China
| | - Lumin Wang
- East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, China
- *Correspondence: Lumin Wang,
| |
Collapse
|
12
|
Yu B, Liu J, Cai Z, Mu T, Gu Y, Xin G, Zhang J. miRNA-mRNA associations with inosine monophosphate specific deposition in the muscle of Jingyuan chicken. Br Poult Sci 2022; 63:821-832. [PMID: 35895079 DOI: 10.1080/00071668.2022.2106777] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
1. Inosine monophosphate (IMP), is an essential component for meat flavour and microRNAs (miRNAs) play a vital role in its post-transcriptional regulation. However, the mechanism of how miRNA expression affects muscle-specific IMP deposition is unclear.2. The following study performed transcriptome sequencing and bioinformatics analysis of breast and leg muscle, which have significantly different IMP content in Jingyuan chicken. The differential miRNA-mRNAs were screened out and correlation analysis with IMP content was performed.3. A total of 39 differentially expressed miRNAs (DE miRNAs) and 666 differentially expressed mRNAs (DE mRNAs) were identified between breast muscles and leg muscles. Using miRNA-mRNA integrated analysis, 29 miRNA-target gene pairs were obtained, composed of 13 DE miRNAs and 28 DE mRNAs. Next, purine metabolism, glycolysis/gluconeogenesis, pyruvate metabolism and the biosynthesis of amino acid pathways as necessary for muscle IMP-specific deposition were identified. The differentially expressed gene PKM2, which was significantly enriched in all four pathways, is involved in IMP anabolism in the form of energy metabolism and enzyme activity regulation. The correlation analysis suggested that the gga-miR-107-3p-KLHDC2 negative interaction may be a key regulator in IMP deposition.4. This study explores the functional mechanism of IMP-specific deposition in Jingyuan chicken muscles at the miRNA and mRNA levels and highlights multiple candidate miRNAs and mRNAs for molecular-assisted breeding.
Collapse
Affiliation(s)
- Baojun Yu
- College of Agriculture, Ningxia University, Yinchuan China
| | - Jiamin Liu
- College of Agriculture, Ningxia University, Yinchuan China
| | - Zhengyun Cai
- College of Agriculture, Ningxia University, Yinchuan China
| | - Tong Mu
- College of Agriculture, Ningxia University, Yinchuan China
| | - Yaling Gu
- College of Agriculture, Ningxia University, Yinchuan China
| | - Guosheng Xin
- School of Life Sciences, Ningxia University/Ningxia Feed Engineering Technology Research Center, Yinchuan China
| | - Juan Zhang
- College of Agriculture, Ningxia University, Yinchuan China
| |
Collapse
|
13
|
Tan K, Wilkinson MF. Regulation of both transcription and RNA turnover contribute to germline specification. Nucleic Acids Res 2022; 50:7310-7325. [PMID: 35776114 PMCID: PMC9303369 DOI: 10.1093/nar/gkac542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/29/2022] [Accepted: 06/29/2022] [Indexed: 12/25/2022] Open
Abstract
The nuanced mechanisms driving primordial germ cells (PGC) specification remain incompletely understood since genome-wide transcriptional regulation in developing PGCs has previously only been defined indirectly. Here, using SLAMseq analysis, we determined genome-wide transcription rates during the differentiation of embryonic stem cells (ESCs) to form epiblast-like (EpiLC) cells and ultimately PGC-like cells (PGCLCs). This revealed thousands of genes undergoing bursts of transcriptional induction and rapid shut-off not detectable by RNAseq analysis. Our SLAMseq datasets also allowed us to infer RNA turnover rates, which revealed thousands of mRNAs stabilized and destabilized during PGCLC specification. mRNAs tend to be unstable in ESCs and then are progressively stabilized as they differentiate. For some classes of genes, mRNA turnover regulation collaborates with transcriptional regulation, but these processes oppose each other in a surprisingly high frequency of genes. To test whether regulated mRNA turnover has a physiological role in PGC development, we examined three genes that we found were regulated by RNA turnover: Sox2, Klf2 and Ccne1. Circumvention of their regulated RNA turnover severely impaired the ESC-to-EpiLC and EpiLC-to-PGCLC transitions. Our study demonstrates the functional importance of regulated RNA stability in germline development and provides a roadmap of transcriptional and post-transcriptional regulation during germline specification.
Collapse
Affiliation(s)
- Kun Tan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Miles F Wilkinson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Institute of Genomic Medicine (IGM), University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
14
|
Post-transcriptional regulation of factors important for the germ line. Curr Top Dev Biol 2022; 146:49-78. [PMID: 35152986 DOI: 10.1016/bs.ctdb.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Echinoderms are a major model system for many general aspects of biology, including mechanisms of gene regulation. Analysis of transcriptional regulation (Gene regulatory networks, direct DNA-binding of proteins to specific cis-elements, and transgenesis) has contributed to our understanding of how an embryo works. This chapter looks at post-transcriptional gene regulation in the context of how the primordial germ cells are formed, and how the factors essential for this process are regulated. Important in echinoderms, as in many embryos, is that key steps of fate determination are made post-transcriptionally. This chapter highlights these steps uncovered in sea urchins and sea stars, and links them to a general theme of how the germ line may regulate its fate differently than many of the embryo's somatic cell lineages.
Collapse
|
15
|
Du S, Zhou L, Wang X, Xu S, Li J, Song Z, Liu Q. Characterization of vasa and dnd homologs in summer flounder, Paralichthys dentatus: Expression analysis and colocalization of PGCs during embryogenesis. Theriogenology 2022; 181:180-189. [PMID: 35121562 DOI: 10.1016/j.theriogenology.2022.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 02/04/2023]
Abstract
Specification of primordial germ cells (PGCs) is particularly important for germline formation. Many maternal-effect genes such as vasa, dnd, and nanos have been identified. However, the research on distribution patterns of PGCs in marine fish is limited. Vasa has been widely used as a germ cell marker to identify its origination in teleosts because vasa RNA is a component of germ plasm. Dnd is known to be an RNA binding protein that protects germline-specific RNAs from degradation. In this study, we isolated full-length vasa and dnd cDNA from summer flounder to track germ cell origination and their expression patterns by RT-PCR and ISH. The results demonstrated that deduced amino acid sequence of Pdvas and Pddnd shared typically conserved motifs of their homologues and demonstrated high identities with other teleosts. Both vasa and dnd transcripts were exclusively detected in germ cells of the gonads. During embryogenesis, vasa and dnd RNA were located at the cleavage furrows of early cleavage stages, and then through proliferation and migration they eventually moved to a location at the predetermined genital ridge. Phylogenetic analysis revealed that summer flounder belongs to the Euteleostei species, but vasa/dnd transcripts localized at the cleavage furrows was similar to that in zebrafish (Osteriophysans). This suggests that germ cells differentiating at early embryogenesis have no direct relation with phylogenesis. At the same time, we found the spatio-temporal expression pattern of dnd was highly consistent with vasa during this process, which indicated the important function of dnd in keeping the target RNA from being degraded to maintain germ cell fate. These results will provide further understanding of germ plasm localization and PGC differentiation in teleosts, and facilitate germ cell manipulation in marine fishes.
Collapse
Affiliation(s)
- Shuran Du
- School of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, 266109, China; CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Li Zhou
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China; College of Life Science, Ningde Normal University, Engineering Research Center of Mindong Aquatic Product Deep-Processing,Fujian Province University, Ningde, 352100, China
| | - Xueying Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shihong Xu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jun Li
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zongcheng Song
- Weihai Shenghang Aquatic Product Science and Technology Co. Ltd., Weihai, 264319, China.
| | - Qinghua Liu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| |
Collapse
|
16
|
Mo C, Li W, Jia K, Liu W, Yi M. Proper Balance of Small GTPase rab10 Is Critical for PGC Migration in Zebrafish. Int J Mol Sci 2021; 22:11962. [PMID: 34769390 PMCID: PMC8584686 DOI: 10.3390/ijms222111962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022] Open
Abstract
MicroRNAs (miRNAs) play important roles in post-transcriptional repression in nearly every biological process including germ cell development. Previously, we have identified a zebrafish germ plasm-specific miRNA miR-202-5p, which regulates PGC migration through targeting cdc42se1 to protect cdc42 expression. However, knockdown of cdc42se1 could not significantly rescue PGC migration in maternal miR-202 mutant (MmiR-202) embryos, indicating that there are other target genes of miR-202-5p required for the regulation of PGC migration. Herein, we revealed the transcriptional profiles of wild type and MmiR-202 PGCs and obtained 129 differentially expressed genes (DEGs), of which 42 DEGs were enriched cell migration-related signaling pathways. From these DEGs, we identified two novel miR-202-5p target genes prdm12b and rab10. Furthermore, we found that disruption of rab10 expression led to significantly migratory defects of PGC by overexpression of rab10 siRNA, or WT, inactive as well as active forms of rab10 mRNA, and WT rab10 overexpression mediated migratory defects could be partially but significantly rescued by overexpression of miR-202-5p, demonstrating that rab10 is an important factor involved miR-202-5p mediated regulation of PGC migration. Taken together, the present results provide significant information for understanding the molecular mechanism by which miR-202-5p regulates PGC migration in zebrafish.
Collapse
Affiliation(s)
- Chengyu Mo
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China; (C.M.); (W.L.); (K.J.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
| | - Wenjing Li
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China; (C.M.); (W.L.); (K.J.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
| | - Kuntong Jia
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China; (C.M.); (W.L.); (K.J.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
| | - Wei Liu
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China; (C.M.); (W.L.); (K.J.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
| | - Meisheng Yi
- School of Marine Sciences, Sun Yat-sen University, Zhuhai 519082, China; (C.M.); (W.L.); (K.J.)
- Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Guangzhou 510275, China
| |
Collapse
|
17
|
Hansen CL, Chamberlain TJ, Trevena RL, Kurek JE, Pelegri F. Conserved germ plasm characteristics across the Danio and Devario lineages. Genesis 2021; 59:e23452. [PMID: 34617657 DOI: 10.1002/dvg.23452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/26/2021] [Accepted: 08/29/2021] [Indexed: 11/06/2022]
Abstract
In many animal species, germ cell specification requires the inheritance of germ plasm, a biomolecular condensate containing maternally derived RNAs and proteins. Most studies of germ plasm composition and function have been performed in widely evolutionarily divergent model organisms, such as Caenorhabditis elegans, Drosophila, Xenopus laevis, and Danio rerio (zebrafish). In zebrafish, 12 RNAs localize to germ plasm at the furrows of the early embryo. Here, we tested for the presence of these RNAs in three additional species within the Danionin clade: Danio kyathit, Danio albolineatus, and Devario aequipinnatus. By visualizing nanos RNA, we find that germ plasm segregation patterns during early embryogenesis are conserved across these species. Ten additional germ plasm RNAs exhibit localization at the furrows of early embryos in all three non-zebrafish Danionin species, consistent with germ plasm localization. One component of zebrafish germ plasm, ca15b, lacked specific localization in embryos of the more distantly related D. aequipinnatus. Our findings show that within a subset of closely related Danionin species, the vast majority of germ plasm RNA components are conserved. At the same time, the lack of ca15b localization in D. aequipinnatus germ plasm highlights the potential for the divergence of germ plasm composition across a restricted phylogenetic space.
Collapse
Affiliation(s)
- Christina L Hansen
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Trevor J Chamberlain
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Ryan L Trevena
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Jacob E Kurek
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, Wisconsin, USA
| | - Francisco Pelegri
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, Wisconsin, USA
| |
Collapse
|
18
|
MicroRNA expression profile in Lampetra morii upon Vibrio anguillarum infection and miR-4561 characterization targeting lip. Commun Biol 2021; 4:995. [PMID: 34417547 PMCID: PMC8379177 DOI: 10.1038/s42003-021-02525-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
As a critical evolutionary pivot between invertebrates and vertebrates, lampreys provide rich genetic information. Lamprey immune protein (LIP) is a key immune regulator. MicroRNAs, well-conserved in the response to immunological stress, remain understudied in lamprey immunity. We generated a lamprey microRNA expression atlas, using deep sequencing, upon Vibrio anguillarum infection. Using comparative methods, we found that miR-4561 potentially regulates innate immunity via interaction with lip. We found a sequence in the 3′-UTR region of LIP mRNA complementary to the miR-4561 seed region; miR-4561 expression was negatively correlated with LIP. During V. anguillarum infection, miR-4561 inhibited LIP expression and bacterial clearance. Notably, LIP expression in supraneural body cells was necessary for the Gram-negative immune response. Additionally, we observed that overexpression of miR-4561 induced apoptosis in embryonic cells, suggesting a role in embryonic development. Collectively, we show lamprey microRNAs may significantly affect gene regulation and provide new insights on LIP-mediated immune regulation. Ma et al characterise differentially expressed miRNAs in Lamprey upon bacterial infection. They identify miR-4561 as being decreased after infection and show that it targets the 3′UTR of the mRNA encoding lamprey immune protein (LIP) involved in lamprey innate immunity and regulates survival in lamprey embryos.
Collapse
|
19
|
Yoon JH, Cho YS, Lee HB, Park JY, Lim HK. Dead-End ( dnd) Gene Cloning and Gonad-Specific Expression Pattern in Starry Flounder ( Platichthys stellatus). Animals (Basel) 2021; 11:2256. [PMID: 34438719 PMCID: PMC8388513 DOI: 10.3390/ani11082256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 11/24/2022] Open
Abstract
dnd is a germline-specific maternal RNA expressed in various vertebrate classes, which encodes an RNA-binding protein that is essential for PGC migration. The purpose of this study is fundamental research about starry flounder dnd gene for germ cell marker development. In this study, we cloned and analyzed the expression levels of Platichthys stellatus dead end (psdnd) in various tissues and embryonic stages. The psdnd gene was isolated from starry flounder ovaries, cloned into a pGEM-t vector, and sequenced. Full-length of psdnd cDNA was 1495 bp long, encoding 395 amino acids. psdnd expression levels were investigated by real-time polymerase chain reaction (qRT-PCR) in various tissues and embryo developmental stages. psdnd transcripts were detected in the testes and ovaries, but not in somatic tissues. Embryonic psdnd expression levels were higher during early embryo development stages than during late embryogenesis; psdnd expression was highest at the 1 cell stage, then gradually decreased throughout the subsequent developmental stages. The spatial expression pattern was analyzed by whole-mount in situ hybridization (WISH). The psdnd transcripts migration pattern was similar with zebrafish (Danio rerio). Our results suggest that psdnd may function as a germ cell-specific marker.
Collapse
Affiliation(s)
- Ji-Hye Yoon
- Department of Interdisciplinary Program of Biomedicine, Health & Life Convergence Science, Mokpo National University (MNU), Muan 58554, Korea; (J.-H.Y.); (H.-B.L.)
| | - Youn-Su Cho
- Department of Fishery Biology, Pukyong National University (PKNU), Busan 48512, Korea;
| | - Hyo-Bin Lee
- Department of Interdisciplinary Program of Biomedicine, Health & Life Convergence Science, Mokpo National University (MNU), Muan 58554, Korea; (J.-H.Y.); (H.-B.L.)
| | - Jung-Yeol Park
- Department of Marine and Fisheries Resources, Mokpo National University (MNU), Muan 58554, Korea;
| | - Han-Kyu Lim
- Department of Interdisciplinary Program of Biomedicine, Health & Life Convergence Science, Mokpo National University (MNU), Muan 58554, Korea; (J.-H.Y.); (H.-B.L.)
| |
Collapse
|
20
|
Integrated Analysis of miR-430 on Steroidogenesis-Related Gene Expression of Larval Rice Field Eel Monopterus albus. Int J Mol Sci 2021; 22:ijms22136994. [PMID: 34209701 PMCID: PMC8269179 DOI: 10.3390/ijms22136994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/17/2021] [Accepted: 06/25/2021] [Indexed: 01/15/2023] Open
Abstract
The present study aims to reveal the mechanism by which miR-430s regulate steroidogenesis in larval rice field eel Monopterus albus. To this end, M. albus embryos were respectively microinjected with miRNA-overexpressing mimics (agomir430a, agomir430b, and agomir430c) or miRNA-knockdown inhibitors (antagomir430a, antagomir430b, and antagomir430c). Transcriptome profiling of the larvae indicated that a total of more than 149 differentially expressed genes (DEGs) were identified among the eight treatments. Specifically, DEGs related to steroidogenesis, the GnRH signaling pathway, the erbB signaling pathway, the Wnt signaling pathway, and other pathways were characterized in the transcriptome. We found that steroidogenesis-related genes (hydroxysteroid 17-beta dehydrogenase 3 (17β-hsdb3), hydroxysteroid 17-beta dehydrogenase 7 (17β-hsdb7), hydroxysteroid 17-beta dehydrogenase 12 (17β-hsdb12), and cytochrome P450 family 19 subfamily a (cyp19a1b)) were significantly downregulated in miR-430 knockdown groups. The differential expressions of miR-430 in three gonads indicated different roles of three miR-430 (a, b, and c) isoforms in regulating steroidogenesis and sex differentiation. Mutation of the miR-430 sites reversed the downregulation of cytochrome P450 family 17 (cyp17), cyp19a1b, and forkhead box L2 (foxl2) reporter activities by miR-430, indicating that miR-430 directly interacted with cyp17, cyp19a1b, and foxl2 genes to inhibit their expressions. Combining these findings, we concluded that miR-430 regulated the steroidogenesis and the biosynthesis of steroid hormones by targeting cyp19a1b in larval M. albus. Our results provide a novel insight into steroidogenesis at the early stage of fish at the molecular level.
Collapse
|
21
|
Baloch AR, Franěk R, Saito T, Pšenička M. Dead-end (dnd) protein in fish-a review. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:777-784. [PMID: 30671782 DOI: 10.1007/s10695-018-0606-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/28/2018] [Indexed: 06/09/2023]
Abstract
Dead end (dnd) is a germ plasm-specific maternal RNA discovered in zebrafish and then in other vertebrates. Dnd protein is essential for migration and motility of primordial germ cells (PGCs), only cells destined to transfer genetic information to offspring. PGCs arise far from somatic cells of developing gonads and they must migrate to their site of function. Migration of PGCs follows complex path by various developing tissues as their disruption impacts on the fertility. Recently, it has been found that dnd is not required for survival of PGCs and dnd-deficient zebrafish PGCs transdifferentiate into the somatic cells. In fish, targeting dnd causes removal of PGCs that ultimately affects sex differentiation. Sterility in various fish species can be achieved by knockdown or knockout of dnd. In our review, we have discussed dnd as a germ cell-specific molecular marker in fish, its interaction with miRNAs, and its use in aquaculture and fish conservation.
Collapse
Affiliation(s)
- Abdul Rasheed Baloch
- South Bohemian Research Center for 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, Zatisi 728/II, 389 25, Vodnany, Czech Republic.
| | - Roman Franěk
- South Bohemian Research Center for 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, Zatisi 728/II, 389 25, Vodnany, Czech Republic
| | - Taiju Saito
- South Bohemian Research Center for 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, Zatisi 728/II, 389 25, Vodnany, Czech Republic
- Nishiura Station, South Ehime Fisheries Research Center, Ehime University, Uchidomari, Ainan, Ehime, 798-4206, Japan
| | - Martin Pšenička
- South Bohemian Research Center for 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, Zatisi 728/II, 389 25, Vodnany, Czech Republic
| |
Collapse
|
22
|
Imarazene B, Beille S, Jouanno E, Branthonne A, Thermes V, Thomas M, Herpin A, Rétaux S, Guiguen Y. Primordial Germ Cell Migration and Histological and Molecular Characterization of Gonadal Differentiation in Pachón Cavefish Astyanax mexicanus. Sex Dev 2021; 14:80-98. [PMID: 33691331 DOI: 10.1159/000513378] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/16/2020] [Indexed: 11/19/2022] Open
Abstract
The genetic regulatory network governing vertebrate gonadal differentiation appears less conserved than previously thought. Here, we investigated the gonadal development of Astyanax mexicanus Pachón cavefish by looking at primordial germ cells (PGCs) migration and proliferation, gonad histology, and gene expression patterns. We showed that PGCs are first detected at the 80% epiboly stage and then reach the gonadal primordium at 1 day post-fertilization (dpf). However, in contrast to the generally described absence of PGCs proliferation during their migration phase, PGCs number in cavefish doubles between early neurula and 8-9 somites stages. Combining both gonadal histology and vasa (germ cell marker) expression patterns, we observed that ovarian and testicular differentiation occurs around 65 dpf in females and 90 dpf in males, respectively, with an important inter-individual variability. The expression patterns of dmrt1, gsdf, and amh revealed a conserved predominant male expression during cavefish gonadal development, but none of the ovarian differentiation genes, i. e., foxl2a, cyp19a1a, and wnt4b displayed an early sexually dimorphic expression, and surprisingly all these genes exhibited predominant expression in adult testes. Altogether, our results lay the foundation for further research on sex determination and differentiation in A. mexicanus and contribute to the emerging picture that the vertebrate sex differentiation downstream regulatory network is less conserved than previously thought, at least in teleost fishes.
Collapse
Affiliation(s)
- Boudjema Imarazene
- INRAE, Laboratoire de Physiologie et Génomique des poissons, Rennes, France.,Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Gif-sur-Yvette, France
| | - Séverine Beille
- INRAE, Laboratoire de Physiologie et Génomique des poissons, Rennes, France
| | - Elodie Jouanno
- INRAE, Laboratoire de Physiologie et Génomique des poissons, Rennes, France
| | - Adéle Branthonne
- INRAE, Laboratoire de Physiologie et Génomique des poissons, Rennes, France
| | - Violette Thermes
- INRAE, Laboratoire de Physiologie et Génomique des poissons, Rennes, France
| | - Manon Thomas
- INRAE, Laboratoire de Physiologie et Génomique des poissons, Rennes, France
| | - Amaury Herpin
- INRAE, Laboratoire de Physiologie et Génomique des poissons, Rennes, France
| | - Sylvie Rétaux
- Université Paris-Saclay, CNRS, Institut des Neurosciences Paris-Saclay, Gif-sur-Yvette, France
| | - Yann Guiguen
- INRAE, Laboratoire de Physiologie et Génomique des poissons, Rennes, France,
| |
Collapse
|
23
|
D'Orazio FM, Balwierz PJ, González AJ, Guo Y, Hernández-Rodríguez B, Wheatley L, Jasiulewicz A, Hadzhiev Y, Vaquerizas JM, Cairns B, Lenhard B, Müller F. Germ cell differentiation requires Tdrd7-dependent chromatin and transcriptome reprogramming marked by germ plasm relocalization. Dev Cell 2021; 56:641-656.e5. [PMID: 33651978 PMCID: PMC7957325 DOI: 10.1016/j.devcel.2021.02.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 10/25/2020] [Accepted: 02/03/2021] [Indexed: 02/09/2023]
Abstract
In many animal models, primordial germ cell (PGC) development depends on maternally deposited germ plasm, which prevents somatic cell fate. Here, we show that PGCs respond to regulatory information from the germ plasm in two distinct phases using two distinct mechanisms in zebrafish. We demonstrate that PGCs commence zygotic genome activation together with the somatic blastocysts with no demonstrable differences in transcriptional and chromatin opening. Unexpectedly, both PGC and somatic blastocysts activate germ-cell-specific genes, which are only stabilized in PGCs by cytoplasmic germ plasm determinants. Disaggregated perinuclear relocalization of germ plasm during PGC migration is regulated by the germ plasm determinant Tdrd7 and is coupled to dramatic divergence between PGC and somatic transcriptomes. This transcriptional divergence relies on PGC-specific cis-regulatory elements characterized by promoter-proximal distribution. We show that Tdrd7-dependent reconfiguration of chromatin accessibility is required for elaboration of PGC fate but not for PGC migration. No evidence for transcriptional activation delay in zebrafish PGCs Germ-plasm-associated post-transcriptional divergence during ZGA Epigenetic reprogramming marks onset of PGC migration Epigenetic reprogramming in PGCs relies on Tdrd7, coupled to germ plasm relocalization
Collapse
Affiliation(s)
- Fabio M D'Orazio
- Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; MRC London Institute of Medical Sciences and Faculty of Medicine, Imperial College, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
| | - Piotr J Balwierz
- Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK; MRC London Institute of Medical Sciences and Faculty of Medicine, Imperial College, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK
| | - Ada Jimenez González
- Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Yixuan Guo
- Department of Oncological Sciences and Huntsman Cancer Institute, Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Lucy Wheatley
- Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Aleksandra Jasiulewicz
- Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Yavor Hadzhiev
- Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Juan M Vaquerizas
- MRC London Institute of Medical Sciences and Faculty of Medicine, Imperial College, London, UK; Max Planck Institute for Molecular Biomedicine, Roentgenstrasse 20, Muenster, Germany
| | - Bradley Cairns
- Department of Oncological Sciences and Huntsman Cancer Institute, Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Boris Lenhard
- MRC London Institute of Medical Sciences and Faculty of Medicine, Imperial College, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College, London, UK.
| | - Ferenc Müller
- Institute of Cancer and Genomics Sciences, Birmingham Centre for Genome Biology, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
24
|
Cabrera-Quio LE, Schleiffer A, Mechtler K, Pauli A. Zebrafish Ski7 tunes RNA levels during the oocyte-to-embryo transition. PLoS Genet 2021; 17:e1009390. [PMID: 33600438 PMCID: PMC7924785 DOI: 10.1371/journal.pgen.1009390] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 03/02/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022] Open
Abstract
Post-transcriptional regulation of gene expression is crucial during the oocyte-to-embryo transition, a highly dynamic process characterized by the absence of nuclear transcription. Thus, changes to the RNA content are solely dependent on RNA degradation. Although several mechanisms that promote RNA decay during embryogenesis have been identified, it remains unclear which machineries contribute to remodeling the maternal transcriptome. Here, we focused on the degradation factor Ski7 in zebrafish. Homozygous ski7 mutant fish had higher proportions of both poor quality eggs and eggs that were unable to develop beyond the one-cell stage. Consistent with the idea that Ski7 participates in remodeling the maternal RNA content, transcriptome profiling identified hundreds of misregulated mRNAs in the absence of Ski7. Furthermore, upregulated genes were generally lowly expressed in wild type, suggesting that Ski7 maintains low transcript levels for this subset of genes. Finally, GO enrichment and proteomic analyses of misregulated factors implicated Ski7 in the regulation of redox processes. This was confirmed experimentally by an increased resistance of ski7 mutant embryos to reductive stress. Our results provide first insights into the physiological role of vertebrate Ski7 as a post-transcriptional regulator during the oocyte-to-embryo transition.
Collapse
Affiliation(s)
| | - Alexander Schleiffer
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| | - Karl Mechtler
- Institute of Molecular Biotechnology, IMBA, Campus Vienna-Biocenter (VBC), Vienna, Austria
| | - Andrea Pauli
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
25
|
Mishima Y, Inoue K. Tethered Function Assay to Study RNA-Regulatory Proteins in Zebrafish Embryos. Methods Mol Biol 2021; 2218:347-354. [PMID: 33606244 DOI: 10.1007/978-1-0716-0970-5_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Many proteins are assumed to mediate post-transcriptional regulation of mRNAs. However, the lack of information about their target mRNAs and functional domains hampers the detailed analysis of their molecular function. Here we describe a method to analyze the post-transcriptional effects of proteins of interest by artificially tethering the protein to a reporter mRNA in zebrafish embryos.
Collapse
Affiliation(s)
- Yuichiro Mishima
- Department of Frontier Life Sciences, Faculty of Lifesciences, Kyoto Sangyo University, Kyoto, Japan.
| | - Kunio Inoue
- Department of Biology, Graduate School of Science, Kobe University, Kobe, Hyogo, Japan
| |
Collapse
|
26
|
Dong YT, Feng HY, Tian XQ, Wang QL, Zhang SF, Ma KY, Qiu GF. Identification of a novel germ cell marker MnTdrd from the oriental river prawn Macrobrachium nipponense. Dev Genes Evol 2020; 231:11-19. [PMID: 33244643 DOI: 10.1007/s00427-020-00671-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/04/2020] [Indexed: 11/25/2022]
Abstract
Germ cell-specific genes play an important role in establishing the reproductive system in sexual organisms and have been used as valuable markers for studying gametogenesis and sex differentiation. Previously, we isolated a vasa transcript as a germ cell marker to trace the origin and migration of germ cells in the oriental river prawn Macrobrachium nipponense. Here, we identified a new germ cell-specific marker MnTdrd RNA and assessed its temporal and spatial expression during oogenesis and embryogenesis. MnTdrd transcripts were expressed in high abundance in unfertilized eggs and embryos at cleavage stage and then dropped significantly during late embryogenesis, suggesting that MnTdrd mRNA is maternally inherited. In situ hybridization of ovarian tissue showed that MnTdrd mRNA was initially present in the cytoplasm of previtellogenic oocyte and localized to the perinuclear region as the accumulation of yolk in vitellogenic oocyte. Whole-mount in situ hybridization of embryos showed that MnTdrd-positive signals were only localized in one blastomere until 16-cell stage. In the blastula, there were approximately 16 MnTdrd-positive blastomeres. During embryonized-zoea stage, the MnTdrd-positive cells aggregated as a cluster and migrated to the genital rudiment which would develop into primordial germ cells (PGCs). The localized expression pattern of MnTdrd transcripts resembled that of the previously identified germ cell marker vasa, supporting the preformation mode of germ cell specification. Therefore, we concluded that MnTdrd, together with vasa, is a component of the germ plasm and might have critical roles in germ cell formation and differentiation in the prawn. Thus, MnTdrd can be used as a novel germ cell marker to trace the origin and migration of germ cells.
Collapse
Affiliation(s)
- Yao-Ting Dong
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
- College of Fisheries and Life Science, Shanghai Ocean University, 999 Huchenghuan Road, Pudong New Area, Shanghai, 201306, China
| | - Hai-Yang Feng
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
- College of Fisheries and Life Science, Shanghai Ocean University, 999 Huchenghuan Road, Pudong New Area, Shanghai, 201306, China
| | - Xiao-Qing Tian
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
- College of Fisheries and Life Science, Shanghai Ocean University, 999 Huchenghuan Road, Pudong New Area, Shanghai, 201306, China
| | - Qi-Liang Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
- College of Fisheries and Life Science, Shanghai Ocean University, 999 Huchenghuan Road, Pudong New Area, Shanghai, 201306, China
| | - Shu-Fang Zhang
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
- College of Fisheries and Life Science, Shanghai Ocean University, 999 Huchenghuan Road, Pudong New Area, Shanghai, 201306, China
| | - Ke-Yi Ma
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
- College of Fisheries and Life Science, Shanghai Ocean University, 999 Huchenghuan Road, Pudong New Area, Shanghai, 201306, China
| | - Gao-Feng Qiu
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China.
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.
- Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China.
- College of Fisheries and Life Science, Shanghai Ocean University, 999 Huchenghuan Road, Pudong New Area, Shanghai, 201306, China.
| |
Collapse
|
27
|
Galagali H, Kim JK. The multifaceted roles of microRNAs in differentiation. Curr Opin Cell Biol 2020; 67:118-140. [PMID: 33152557 DOI: 10.1016/j.ceb.2020.08.015] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
MicroRNAs (miRNAs) are major drivers of cell fate specification and differentiation. The post-transcriptional regulation of key molecular factors by microRNAs contributes to the progression of embryonic and postembryonic development in several organisms. Following the discovery of lin-4 and let-7 in Caenorhabditis elegans and bantam microRNAs in Drosophila melanogaster, microRNAs have emerged as orchestrators of cellular differentiation and developmental timing. Spatiotemporal control of microRNAs and associated protein machinery can modulate microRNA activity. Additionally, adaptive modulation of microRNA expression and function in response to changing environmental conditions ensures that robust cell fate specification during development is maintained. Herein, we review the role of microRNAs in the regulation of differentiation during development.
Collapse
Affiliation(s)
- Himani Galagali
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - John K Kim
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
28
|
Zhou L, Wang X, Du S, Wang Y, Zhao H, Du T, Yu J, Wu L, Song Z, Liu Q, Li J. Germline Specific Expression of a vasa Homologue Gene in the Viviparous Fish Black Rockfish ( Sebastes schlegelii) and Functional Analysis of the vasa 3 ' Untranslated Region. Front Cell Dev Biol 2020; 8:575788. [PMID: 33330452 PMCID: PMC7732447 DOI: 10.3389/fcell.2020.575788] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/18/2020] [Indexed: 11/13/2022] Open
Abstract
Germ cells play a key role in gonad development. As precursors, primordial germ cells (PGCs) are particularly important for germline formation. However, the origination and migration patterns of PGCs are poorly studied in marine fish, especially for viviparous economic species. The vasa gene has been widely used as a germ cell marker to identify a germline because vasa RNA is a component of germ plasm. In this study, we described the expression pattern of black rockfish (Sebastes schlegelii) vasa (Ssvas) in gonadal formation and development by in situ hybridization. The results showed that Ssvas failed in localization at the cleavage furrows until the late gastrula stage, when PGCs appeared and migrated to the genital ridge and formed elongated gonadal primordia at 10 days after birth. This study firstly revealed the PGCs origination and migration characteristics in viviparous marine fish. Furthermore, we microinjected chimeric mRNA containing EGFP and the 3′untranslated region (3′UTR) of Ssvas into zebrafish (Danio rerio) and marine medaka (Oryzias melastigma) fertilized eggs for tracing PGCs. We found that, although Sebastes schlegelii lacked early localization, similar to red seabream (Pagrus major) and marine medaka, only the 3′UTR of Ssvas vasa 3′UTR of black rockfish was able to label both zebrafish and marine medaka PGCs. In comparison with other three Euteleostei species, besides some basal motifs, black rockfish had three specific motifs of M10, M12, and M19 just presented in zebrafish, which might play an important role in labeling zebrafish PGCs. These results will promote germ cell manipulation technology development and facilitate artificial reproduction regulation in aquaculture.
Collapse
Affiliation(s)
- Li Zhou
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xueying Wang
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Shuran Du
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yanfeng Wang
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Haixia Zhao
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Tengfei Du
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiachen Yu
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lele Wu
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zongcheng Song
- Weihai Shenghang Aquatic Product Science and Technology Co., Ltd., Weihai, China
| | - Qinghua Liu
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jun Li
- The Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
29
|
Chang CT, Lee YH, HuangFu WC, Liu IH. Cell-intrinsic Fgf signaling contributes to primordial germ cell homing in zebrafish. Theriogenology 2020; 158:424-431. [PMID: 33039926 DOI: 10.1016/j.theriogenology.2020.09.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022]
Abstract
Primordial germ cells (PGCs) are specified before gastrulation and migrate toward the developing gonads. Previous in vitro studies have demonstrated a cell-intrinsic requirement of fibroblast growth factors (FGFs) by PGCs; however, no evidence suggests FGFs signal directly to PGCs in vivo. Here, using zebrafish as the animal model, we identified the mRNA expressions of Fgf receptors (Fgfrs) and determined the roles of Fgf signaling in migrating PGCs. To clarify the functions of Fgf signaling, we manipulated Fgf signaling specifically in PGCs using dominant-negative (dn) and constitutively-active (ca) Fgfrs and revealed a requirement of a basal Fgf signaling level for the robust arrival of PGCs. Repression of Fgf signaling in PGCs swayed the marginal positioning of PGCs as early as 6 h post-fertilization (6 hpf) and disrupted their arrival at the gonadal ridge at 24 hpf. On the other hand, the ectopic PGC phenotypes caused by the dn-Fgfrs could be alleviated by constitutive activation of Fgf signaling. In addition, we carefully ruled out the somatic effects in mosaic embryos by injecting RNA materials into one blastomere of the four- or eight-cell stage embryos. Injection of dn-Fgfrs into one of eight blastomeres hampered the arrival of only the treated PGCs, while the other PGCs remained unaffected. Furthermore, mosaic treatment of ca-Fgfrs rescued the ectopic rates of dn-Fgfr treated PGCs, while the other PGCs remained more ectopic within the same embryos. Interestingly, PGC-specific repression of Fgf signaling did not compromise the PGC number. To our knowledge, this is the first in vivo evidence to show that Fgf signaling plays a cell-intrinsic role in the migration of vertebrate PGCs.
Collapse
Affiliation(s)
- Chia-Teng Chang
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
| | - Yen-Hua Lee
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan
| | - Wei-Chun HuangFu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei, 106, Taiwan; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, 106, Taiwan; School of Veterinary Medicine, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
30
|
Ge S, Dan C, Xiong Y, Gong G, Mei J, Guo W, Li X. Identifying difference in primordial germ cells between XX female and XY male yellow catfish embryos. Gene 2020; 761:145037. [PMID: 32777526 DOI: 10.1016/j.gene.2020.145037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 08/02/2020] [Accepted: 08/04/2020] [Indexed: 11/16/2022]
Abstract
Primordial germ cells (PGCs) are singled out from somatic cells very early during embryogenesis, then they migrate towards the genital ridge and differentiate into gametes through oogenesis or spermatogenesis. Labeling PGCs with Localized RNAexpression (LRE) technique by fluorescent proteins has been widely applied among teleost species to study the germ cell development and gonad differentiation. In this study, we first cloned and characterized the 3' untranslated regions (3'UTRs) of nanos homolog 1-like (nos1l), dead end (dnd), and vasa in yellow catfish (Pelteobagrus fulvidraco), and then synthesized the GFP-nos1l/dnd/vasa 3'UTR mRNAs. Each of these three 3'UTRs could label PGCs in yellow catfish embryos, of which, vasa 3'UTR exhibited the highest labeling efficiency. To identify the differences in PGCs at embryonic stage, XX all-female and XY all-male yellow catfish embryos were produced and injected with GFP-vasa 3'UTR mRNA. We observed the PGC migration route in these two monosex embryos from 24 hpf to 7 dpf, and found there was no difference between them. Besides, the PGC number was counted at 48 hpf, and the result showed that the average PGC number in XX females (11.3) was significantly larger than that in XY males (8.1).These findings provide an insight into the development of PGCs in yellow catfish embryos and the relationship between embryonicPGCnumberand thelatergonaddifferentiation.
Collapse
Affiliation(s)
- Si Ge
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Cheng Dan
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430070, China
| | - Yang Xiong
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Gaorui Gong
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jie Mei
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjie Guo
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Xiaohui Li
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
31
|
Liu Y, Zhu Z, Ho IHT, Shi Y, Li J, Wang X, Chan MTV, Cheng CHK. Genetic Deletion of miR-430 Disrupts Maternal-Zygotic Transition and Embryonic Body Plan. Front Genet 2020; 11:853. [PMID: 32849832 PMCID: PMC7417628 DOI: 10.3389/fgene.2020.00853] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/13/2020] [Indexed: 11/13/2022] Open
Abstract
MiR-430 is considered an important regulator during embryonic development, but genetic loss-of-function study is still lacking. Here we demonstrated that genetic deletion of the miR-430 cluster resulted in developmental defects in cell movement, germ layer specification, axis patterning and organ progenitor formation in zebrafish. Transcriptome analysis indicated that the maternally provided transcripts were not properly degraded whereas the zygotic genome expressed genes were not fully activated in the miR-430 mutants. We further found that a reciprocal regulatory loop exists between miR-430 and maternally provided transcripts: the maternally provided transcripts (Nanog, Dicer1, Dgcr8, and AGOs) are required for miR-430 biogenesis and function, whereas miR-430 is required for the clearance of these maternally provided transcripts. These data provide the first genetic evidence that miR-430 is required for maternal-zygotic transition and subsequent establishment of embryonic body plan.
Collapse
Affiliation(s)
- Yun Liu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Shenzhen Zelong Biological Technology Limited Cooperation, Shenzhen, China
| | - Zeyao Zhu
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
| | - Idy H T Ho
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Department of Anesthesia and Intensive Care, Prince of Wales Hospital, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yujian Shi
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Xia Wang
- Department of Anatomy, Histology and Developmental Biology, School of Basic Medical Sciences, Shenzhen University, Shenzhen, China
| | - Matthew T V Chan
- Department of Anesthesia and Intensive Care, Prince of Wales Hospital, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
32
|
Jin Y, Liu W, Xiang Y, Zhang W, Zhang H, Jia K, Yi M. Maternal miR-202-5p is required for zebrafish primordial germ cell migration by protecting small GTPase Cdc42. J Mol Cell Biol 2020; 12:530-542. [PMID: 31742346 PMCID: PMC7493028 DOI: 10.1093/jmcb/mjz103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 06/10/2019] [Accepted: 07/29/2019] [Indexed: 01/17/2023] Open
Abstract
In many lower animals, germ cell formation, migration, and maintenance depend on maternally provided determinants in germ plasm. In zebrafish, these processes have been extensively studied in terms of RNA-binding proteins and other coding genes. The role of small non-coding RNAs in the regulation of primordial germ cell (PGC) development remains largely unknown and poorly investigated, even though growing interests for the importance of miRNAs involved in a wide variety of biological processes. Here, we reported the role and mechanism of the germ plasm-specific miRNA miR-202-5p in PGC migration: (i) both maternal loss and knockdown of miR-202-5p impaired PGC migration indicated by the mislocalization and reduced number of PGCs; (ii) cdc42se1 was a direct target gene of miR-202-5p, and overexpression of Cdc42se1 in PGCs caused PGC migration defects similar to those observed in loss of miR-202-5p mutants; (iii) Cdc42se1 not only interacted with Cdc42 but also inhibited cdc42 transcription, and overexpression of Cdc42 could rescue PGC migration defects in Cdc42se1 overexpressed embryos. Thus, miR-202-5p regulates PGC migration by directly targeting and repressing Cdc42se1 to protect the expression of Cdc42, which interacts with actin to direct PGC migration.
Collapse
Affiliation(s)
- Yilin Jin
- School of Marine Sciences Sun Yat-sen University, Guangzhou 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 519082, China
| | - Wei Liu
- School of Marine Sciences Sun Yat-sen University, Guangzhou 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 519082, China
| | - Yangxi Xiang
- School of Marine Sciences Sun Yat-sen University, Guangzhou 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 519082, China
| | - Wanwan Zhang
- School of Marine Sciences Sun Yat-sen University, Guangzhou 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 519082, China
| | - Hong Zhang
- School of Marine Sciences Sun Yat-sen University, Guangzhou 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 519082, China
| | - Kuntong Jia
- School of Marine Sciences Sun Yat-sen University, Guangzhou 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 519082, China
- Correspondence to: Meisheng Yi, E-mail: ; Kuntong Jia, E-mail:
| | - Meisheng Yi
- School of Marine Sciences Sun Yat-sen University, Guangzhou 519082, China
- Guangdong Provincial Key Laboratory of Marine Resources and Coastal Engineering, Sun Yat-sen University, Guangzhou 519082, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Sun Yat-sen University, Guangzhou 519082, China
- Correspondence to: Meisheng Yi, E-mail: ; Kuntong Jia, E-mail:
| |
Collapse
|
33
|
Moravec CE, Pelegri F. The role of the cytoskeleton in germ plasm aggregation and compaction in the zebrafish embryo. Curr Top Dev Biol 2020; 140:145-179. [PMID: 32591073 DOI: 10.1016/bs.ctdb.2020.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The transmission of genetic information from one generation to another is crucial for survival of animal species. This is accomplished by the induction of primordial germ cells (PGCs) that will eventually establish the germline. In some animals the germline is induced by signals in gastrula, whereas in others it is specified by inheritance of maternal determinants, known as germ plasm. In zebrafish, aggregation and compaction of maternally derived germ plasm during the first several embryonic cell cycles is essential for generation of PGCs. These processes are controlled by cellular functions associated with the cellular division apparatus. Ribonucleoparticles containing germ plasm components are bound to both the ends of astral microtubules and a dynamic F-actin network through a mechanism integrated with that which drives the cell division program. In this chapter we discuss the role that modifications of the cell division apparatus, including the cytoskeleton and cytoskeleton-associated proteins, play in the regulation of zebrafish germ plasm assembly.
Collapse
Affiliation(s)
- Cara E Moravec
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, United States
| | - Francisco Pelegri
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
34
|
Ahkin Chin Tai JK, Freeman JL. Zebrafish as an integrative vertebrate model to identify miRNA mechanisms regulating toxicity. Toxicol Rep 2020; 7:559-570. [PMID: 32373477 PMCID: PMC7195498 DOI: 10.1016/j.toxrep.2020.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/22/2020] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
Zebrafish are an established vertebrate model for toxicity studies. Zebrafish have a fully sequenced genome and the capability to create genetic models. Zebrafish have over 80 % homology for genes related to human disease. Functions of miRNAs in the zebrafish genome are being characterized. Zebrafish are ideal for mechanistic studies on how miRNAs regulate toxicity.
Zebrafish (Danio rerio) are an integrative vertebrate model ideal for toxicity studies. The zebrafish genome is sequenced with detailed characterization of all life stages. With their genetic similarity to humans, zebrafish models are established to study biological processes including development and disease mechanisms for translation to human health. The zebrafish genome, similar to other eukaryotic organisms, contains microRNAs (miRNAs) which function along with other epigenetic mechanisms to regulate gene expression. Studies have now established that exposure to toxins and xenobiotics can change miRNA expression profiles resulting in various physiological and behavioral alterations. In this review, we cover the intersection of miRNA alterations from toxin or xenobiotic exposure with a focus on studies using the zebrafish model system to identify miRNA mechanisms regulating toxicity. Studies to date have addressed exposures to toxins, particulate matter and nanoparticles, various environmental contaminants including pesticides, ethanol, and pharmaceuticals. Current limitations of the completed studies and future directions for this research area are discussed.
Collapse
Affiliation(s)
| | - Jennifer L Freeman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907 USA
| |
Collapse
|
35
|
Yang F, Qi J. miR-430a regulates the development of left-right asymmetry by targeting sqt in the teleost. Gene 2020; 745:144628. [PMID: 32224271 DOI: 10.1016/j.gene.2020.144628] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 03/24/2020] [Accepted: 03/25/2020] [Indexed: 10/24/2022]
Abstract
microRNAs (miRNAs) are short, endogenous non-coding RNAs that contain approximately 18-22 nucleotides. miRNAs are involved in gene regulation by recognizing and binding the 3'UTR of target gene. In our previous data, miR-430 family showed significant differential expression modes through metamorphosis in Japanese flounder. It was speculated that miR-430a plays a key role in left-right patterning. We predicted the targets of miR-430a and gene ontology (GO) was performed. We speculated miR-430a is involved in the basal molecular function and organ development. In Japanese flounder, sqt as a target of miR-430a was enriched into heart development term. Sqt has been reported to participate in mesendoderm formation and organ development. Cardiac morphogenesis is the first asymmetric development process, which breaks left-right symmetry in bilateria. It was used as a marker to detect L-R asymmetric effects of miR-430a. Overexpression and suppression of miR-430a resulted in abnormal KV (Kupffer's vesicles) development and disordered in nodal-related expression with consequent cardiac laterality. Squint mRNA of Japanese flounder (Posqt) as a target of miR-430a was overexpressed and caused similar phenotype with miR-430a suppression group, such as longer cilia in KV and high range of clmc2 and spaw ectopic expression. Moreover, rescue experiments were performed and suggested that cardiac and KV defections, induced by overexpressing miR-430a, could be rescued by injecting Posqt mRNA. These results suggested that miR-430a regulates the development of left-right asymmetry by targeting sqt in the teleost.
Collapse
Affiliation(s)
- Fan Yang
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China
| | - Jie Qi
- MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, 5 Yushan Road, Qingdao 266003, China.
| |
Collapse
|
36
|
Westerich KJ, Chandrasekaran KS, Gross-Thebing T, Kueck N, Raz E, Rentmeister A. Bioorthogonal mRNA labeling at the poly(A) tail for imaging localization and dynamics in live zebrafish embryos. Chem Sci 2020; 11:3089-3095. [PMID: 33623655 PMCID: PMC7879197 DOI: 10.1039/c9sc05981d] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/21/2020] [Indexed: 12/14/2022] Open
Abstract
Live imaging of mRNA in cells and organisms is important for understanding the dynamic aspects underlying its function.
Live imaging of mRNA in cells and organisms is important for understanding the dynamic aspects underlying its function. Ideally, labeling of mRNA should not alter its structure or function, nor affect the biological system. However, most methods applied in vivo make use of genetically encoded tags and reporters that significantly enhance the size of the mRNA of interest. Alternately, we utilize the 3′ poly(A) tail as a non-coding repetitive hallmark to covalently label mRNAs via bioorthogonal chemistry with different fluorophores from a wide range of spectra without significantly changing the size. We demonstrate that the labeled mRNAs can be visualized in cells and zebrafish embryos, and that they are efficiently translated. Importantly, the labeled mRNAs acquired the proper subcellular localization in developing zebrafish embryos and their dynamics could be tracked in vivo.
Collapse
Affiliation(s)
- Kim J Westerich
- Institute of Cell Biology Center for Molecular Biology of Inflammation , University of Münster , D-48149 Münster , Germany .
| | - Karthik S Chandrasekaran
- Institut für Biochemie , Westfälische Wilhelms-Universität Münster , Wilhelm-Klemm-Str. 2 , 48149 Münster , Germany .
| | - Theresa Gross-Thebing
- Institute of Cell Biology Center for Molecular Biology of Inflammation , University of Münster , D-48149 Münster , Germany .
| | - Nadine Kueck
- Institut für Biochemie , Westfälische Wilhelms-Universität Münster , Wilhelm-Klemm-Str. 2 , 48149 Münster , Germany .
| | - Erez Raz
- Cells in Motion Interfaculty Centre (CiMIC) , Waldeyerstraße 15 , D-48149 Münster , Germany.,Institute of Cell Biology Center for Molecular Biology of Inflammation , University of Münster , D-48149 Münster , Germany .
| | - Andrea Rentmeister
- Cells in Motion Interfaculty Centre (CiMIC) , Waldeyerstraße 15 , D-48149 Münster , Germany.,Institut für Biochemie , Westfälische Wilhelms-Universität Münster , Wilhelm-Klemm-Str. 2 , 48149 Münster , Germany .
| |
Collapse
|
37
|
Gross-Thebing T, Raz E. Dead end and Detour: The function of the RNA-binding protein Dnd in posttranscriptional regulation in the germline. Curr Top Dev Biol 2020; 140:181-208. [DOI: 10.1016/bs.ctdb.2019.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Zebrafish embryogenesis – A framework to study regulatory RNA elements in development and disease. Dev Biol 2020; 457:172-180. [DOI: 10.1016/j.ydbio.2019.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 12/26/2022]
|
39
|
Balasubramanian S, Raghunath A, Perumal E. Role of epigenetics in zebrafish development. Gene 2019; 718:144049. [DOI: 10.1016/j.gene.2019.144049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
|
40
|
Zhou L, Wang X, Liu Q, Xu S, Zhao H, Han M, Wang Y, Song Z, Li J. Visualization of Turbot (Scophthalmus maximus) Primordial Germ Cells in vivo Using Fluorescent Protein Mediated by the 3' Untranslated Region of nanos3 or vasa Gene. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2019; 21:671-682. [PMID: 31502176 DOI: 10.1007/s10126-019-09911-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 07/03/2019] [Indexed: 06/10/2023]
Abstract
Primordial germ cells (PGCs) as the precursors of germ cells are responsible for transmitting genetic information to the next generation. Visualization of teleost PGCs in vivo is essential to research the origination and development of germ cells and facilitate further manipulation on PGCs isolation, cryopreservation, and surrogate breeding. In this study, artificially synthesized mRNAs constructed by fusing fluorescent protein coding region to the 3' untranslated region (3'UTR) of nanos3 or vasa (mCherry-Smnanos3 3'UTR or mCherry-Smvasa 3'UTR mRNA) were injected into turbot (Scophthalmus maximus) fertilized eggs for tracing PGCs. The results demonstrated that the fluorescent PGCs differentiated from somatic cells and aligned on both sides of the trunk at the early segmentation period, then migrated and located at the dorsal part of the gut where the gonad would form. In the same way, we also found that the zebrafish (Danio rerio) vasa 3'UTR could trace turbot PGCs, while the vasa 3'UTR s of marine medaka (Oryzias melastigma) and red seabream (Pagrus major) failed, although they could label the marine medaka PGCs. In addition, through comparative analysis, we discovered that some potential sequence elements in the3 'UTRs of nanos3 and vasa, such as GCACs, 62-bp U-rich regions and nucleotide 187-218 regions might be involved in PGCs stabilization. The results of this study provided an efficient, rapid, and specific non-transgenic approach for visualizing PGCs of economical marine fish in vivo.
Collapse
Affiliation(s)
- Li Zhou
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xueying Wang
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
| | - Qinghua Liu
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, P. R. China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China.
| | - Shihong Xu
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
| | - Haixia Zhao
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mingming Han
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
| | - Yunong Wang
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, P. R. China
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zongcheng Song
- Weihai Shenghang Aquatic Product Science and Technology Co. Ltd., Weihai, 264200, China
| | - Jun Li
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China.
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, 7 Nanhai Road, Qingdao, 266071, P. R. China.
- Center for Ocean Mega-Science, Chinese Academy of Sciences, 7 Nanhai Road, Qingdao, 266071, P. R. China.
| |
Collapse
|
41
|
Nunez L, Mokkapati S, Yu C, Deng JM, Behringer RR, Matin A. Generation of a novel mouse strain with conditional, cell-type specific, expression of DND1. Genesis 2019; 57:e23335. [PMID: 31513344 DOI: 10.1002/dvg.23335] [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: 04/11/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 01/26/2023]
Abstract
Dead-End 1 (DND1) encodes an RNA binding protein critical for viable primordial germ cells in vertebrates. When introduced into cancer cell lines, DND1 suppresses cell proliferation and enhances apoptosis. However, the molecular function of mammalian wild-type DND1 has mostly been studied in cell lines and not verified in the organism. To facilitate study of wild-type DND1 function in mammalian systems, we generated a novel transgenic mouse line, LSL-FM-DND1 flox/+ , which conditionally expresses genetically engineered, FLAG-tagged and myc-tagged DND1 in a cell type-specific manner. We report that FLAG-myc-DND1 is indeed expressed in specific tissues of the mouse when LSL-FM-DND1 flox/+ is combined with mouse strains expressing Cre-recombinase. LSL-FM-DND1 flox/+ mice are fertile with no overt health effects. We expressed FLAG-myc-DND1 in the pancreas and found that chronic, ectopic expression of FLAG-myc-DND1 led to increase in fasting glucose levels in older mice. Thus, this novel LSL-FM-DND1 flox/+ mouse strain will facilitate studies on the biological and molecular function of wild-type DND1.
Collapse
Affiliation(s)
- Lisa Nunez
- Department of Pharmaceutical Sciences, Texas Southern University, Houston, Texas
| | - Sharada Mokkapati
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chengtai Yu
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Jian M Deng
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Richard R Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Angabin Matin
- Department of Pharmaceutical Sciences, Texas Southern University, Houston, Texas
| |
Collapse
|
42
|
Fischer P, Chen H, Pacho F, Rieder D, Kimmel RA, Meyer D. FoxH1 represses miR-430 during early embryonic development of zebrafish via non-canonical regulation. BMC Biol 2019; 17:61. [PMID: 31362746 PMCID: PMC6664792 DOI: 10.1186/s12915-019-0683-z] [Citation(s) in RCA: 5] [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: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND FoxH1 is a forkhead transcription factor with conserved key functions in vertebrate mesoderm induction and left-right patterning downstream of the TGF-beta/Nodal signaling pathway. Binding of the forkhead domain (FHD) of FoxH1 to a highly conserved proximal sequence motif was shown to regulate target gene expression. RESULTS We identify the conserved microRNA-430 family (miR-430) as a novel target of FoxH1. miR-430 levels are increased in foxH1 mutants, resulting in a reduced expression of transcripts that are targeted by miR-430 for degradation. To determine the underlying mechanism of miR-430 repression, we performed chromatin immunoprecipitation studies and overexpression experiments with mutant as well as constitutive active and repressive forms of FoxH1. Our studies reveal a molecular interaction of FoxH1 with miR-430 loci independent of the FHD. Furthermore, we show that previously described mutant forms of FoxH1 that disrupt DNA binding or that lack the C-terminal Smad Interaction Domain (SID) dominantly interfere with miR-430 repression, but not with the regulation of previously described FoxH1 targets. CONCLUSIONS We were able to identify the distinct roles of protein domains of FoxH1 in the regulation process of miR-430. We provide evidence that the indirect repression of miR-430 loci depends on the connection to a distal repressive chromosome environment via a non-canonical mode. The widespread distribution of such non-canonical binding sites of FoxH1, found not only in our study, argues against a function restricted to regulating miR-430 and for a more global role of FoxH1 in chromatin folding.
Collapse
Affiliation(s)
- Patrick Fischer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Hao Chen
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Frederic Pacho
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Dietmar Rieder
- Division of Bioinformatics, Biocenter, Innsbruck Medical University, Innrain 80, 6020, Innsbruck, Austria
| | - Robin A Kimmel
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Dirk Meyer
- Institute of Molecular Biology/CMBI, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria.
| |
Collapse
|
43
|
Skvortsova K, Tarbashevich K, Stehling M, Lister R, Irimia M, Raz E, Bogdanovic O. Retention of paternal DNA methylome in the developing zebrafish germline. Nat Commun 2019; 10:3054. [PMID: 31296860 PMCID: PMC6624265 DOI: 10.1038/s41467-019-10895-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/31/2019] [Indexed: 01/08/2023] Open
Abstract
Two waves of DNA methylation reprogramming occur during mammalian embryogenesis; during preimplantation development and during primordial germ cell (PGC) formation. However, it is currently unclear how evolutionarily conserved these processes are. Here we characterise the DNA methylomes of zebrafish PGCs at four developmental stages and identify retention of paternal epigenetic memory, in stark contrast to the findings in mammals. Gene expression profiling of zebrafish PGCs at the same developmental stages revealed that the embryonic germline is defined by a small number of markers that display strong developmental stage-specificity and that are independent of DNA methylation-mediated regulation. We identified promoters that are specifically targeted by DNA methylation in somatic and germline tissues during vertebrate embryogenesis and that are frequently misregulated in human cancers. Together, these detailed methylome and transcriptome maps of the zebrafish germline provide insight into vertebrate DNA methylation reprogramming and enhance our understanding of the relationships between germline fate acquisition and oncogenesis.
Collapse
Affiliation(s)
- Ksenia Skvortsova
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Katsiaryna Tarbashevich
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, 48149, Germany
| | - Martin Stehling
- Flow Cytometry Unit, Max-Planck-Institute for Molecular Biomedicine, Roentgenstraße 20, 48149, Münster, Germany
| | - Ryan Lister
- ARC CoE Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, WA, 6009, Australia
- Molecular Medicine Division, Harry Perkins Institute of Medical Research, Perth, WA, 6009, Australia
| | - Manuel Irimia
- Centre for Genomic Regulation, The Barcelona Institute for Science and Technology, Barcelona, 08003, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, 08002, Spain
- ICREA, Barcelona, 08010, Spain
| | - Erez Raz
- Institute of Cell Biology, Center for Molecular Biology of Inflammation, University of Münster, Münster, 48149, Germany
| | - Ozren Bogdanovic
- Genomics and Epigenetics Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia.
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2010, Australia.
| |
Collapse
|
44
|
Zaucker A, Nagorska A, Kumari P, Hecker N, Wang Y, Huang S, Cooper L, Sivashanmugam L, VijayKumar S, Brosens J, Gorodkin J, Sampath K. Translational co-regulation of a ligand and inhibitor by a conserved RNA element. Nucleic Acids Res 2019; 46:104-119. [PMID: 29059375 PMCID: PMC5758872 DOI: 10.1093/nar/gkx938] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 10/03/2017] [Indexed: 12/20/2022] Open
Abstract
In many organisms, transcriptional and post-transcriptional regulation of components of pathways or processes has been reported. However, to date, there are few reports of translational co-regulation of multiple components of a developmental signaling pathway. Here, we show that an RNA element which we previously identified as a dorsal localization element (DLE) in the 3'UTR of zebrafish nodal-related1/squint (ndr1/sqt) ligand mRNA, is shared by the related ligand nodal-related2/cyclops (ndr2/cyc) and the nodal inhibitors, lefty1 (lft1) and lefty2 mRNAs. We investigated the activity of the DLEs through functional assays in live zebrafish embryos. The lft1 DLE localizes fluorescently labeled RNA similarly to the ndr1/sqt DLE. Similar to the ndr1/sqt 3'UTR, the lft1 and lft2 3'UTRs are bound by the RNA-binding protein (RBP) and translational repressor, Y-box binding protein 1 (Ybx1), whereas deletions in the DLE abolish binding to Ybx1. Analysis of zebrafish ybx1 mutants shows that Ybx1 represses lefty1 translation in embryos. CRISPR/Cas9-mediated inactivation of human YBX1 also results in human NODAL translational de-repression, suggesting broader conservation of the DLE RNA element/Ybx1 RBP module in regulation of Nodal signaling. Our findings demonstrate translational co-regulation of components of a signaling pathway by an RNA element conserved in both sequence and structure and an RBP, revealing a 'translational regulon'.
Collapse
Affiliation(s)
- Andreas Zaucker
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Agnieszka Nagorska
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Pooja Kumari
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Nikolai Hecker
- Center for non-coding RNAs in Technology and Health, Department of Veterinary and Animal Sciences, Faculty for Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 3, 1870 Frederiksberg C, Denmark
| | - Yin Wang
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Sizhou Huang
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Ledean Cooper
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Lavanya Sivashanmugam
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Shruthi VijayKumar
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Jan Brosens
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| | - Jan Gorodkin
- Center for non-coding RNAs in Technology and Health, Department of Veterinary and Animal Sciences, Faculty for Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 3, 1870 Frederiksberg C, Denmark
| | - Karuna Sampath
- Cell & Developmental Biology Unit, Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
45
|
Guo H, Du X, Zhang Y, Wu J, Wang C, Li M, Hua X, Zhang XA, Yan J. Specific miRNA-G Protein-Coupled Receptor Networks Regulate Sox9a/Sox9b Activities to Promote Gonadal Rejuvenation in Zebrafish. Stem Cells 2019; 37:1189-1199. [DOI: 10.1002/stem.3040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/17/2019] [Accepted: 05/04/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Huiping Guo
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Xinlu Du
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Ying Zhang
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Jiacheng Wu
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Chenghui Wang
- Department of Aquaculture; Shanghai Ocean University; Lingang New City, Shanghai People's Republic of China
| | - Mingyou Li
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources; Ministry of Education; Shanghai People's Republic of China
| | - Xianxin Hua
- Department of Cancer Biology; University of Pennsylvania Perelman School of Medicine; Philadelphia, Pennsylvania USA
| | - Xin A. Zhang
- Stephenson Cancer Center and Department of Physiology; The University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma USA
| | - Jizhou Yan
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources; Ministry of Education; Shanghai People's Republic of China
| |
Collapse
|
46
|
A state-of-the-art review of surrogate propagation in fish. Theriogenology 2019; 133:216-227. [DOI: 10.1016/j.theriogenology.2019.03.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 03/30/2019] [Indexed: 12/20/2022]
|
47
|
Vastenhouw NL, Cao WX, Lipshitz HD. The maternal-to-zygotic transition revisited. Development 2019; 146:146/11/dev161471. [PMID: 31189646 DOI: 10.1242/dev.161471] [Citation(s) in RCA: 261] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The development of animal embryos is initially directed by maternal gene products. Then, during the maternal-to-zygotic transition (MZT), developmental control is handed to the zygotic genome. Extensive research in both vertebrate and invertebrate model organisms has revealed that the MZT can be subdivided into two phases, during which very different modes of gene regulation are implemented: initially, regulation is exclusively post-transcriptional and post-translational, following which gradual activation of the zygotic genome leads to predominance of transcriptional regulation. These changes in the gene expression program of embryos are precisely controlled and highly interconnected. Here, we review current understanding of the mechanisms that underlie handover of developmental control during the MZT.
Collapse
Affiliation(s)
- Nadine L Vastenhouw
- Max Planck Institute of Molecular Cell Biology and Genetics, Pfotenhauerstraße 108, 01307 Dresden, Germany
| | - Wen Xi Cao
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| | - Howard D Lipshitz
- Department of Molecular Genetics, University of Toronto, 661 University Avenue, Toronto, Ontario M5G 1M1, Canada
| |
Collapse
|
48
|
Xiao Q, Sun Y, Liang X, Zhang L, Onxayvieng K, Li Z, Li D. Visualizing primordial germ cell migration in embryos of rice field eel (Monopterus albus) using fluorescent protein tagged 3' untranslated regions of nanos3, dead end and vasa. Comp Biochem Physiol B Biochem Mol Biol 2019; 235:62-69. [PMID: 31176867 DOI: 10.1016/j.cbpb.2019.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/25/2019] [Accepted: 06/03/2019] [Indexed: 11/17/2022]
Abstract
In rice field eel (Monopterus albus), germ cell development in the developing gonad has been revealed in detail. However, it is unclear how primordial germ cells (PGCs) migrate to the somatic part of the gonad (genital ridge). This study visualized PGC migration by injecting a chimeric mRNA containing a fluorescent protein fused to the 3' untranslated region (3'UTR) of three different genes, nanos3 of zebrafish (Danio rerio) and dead end (dnd) and vasa of rice field eel. The mRNAs were injected either alone or in pairs into embryos at the one-cell stage. The results showed that mRNAs containing nanos3 and dnd 3'UTRs labeled PGCs over a wider time frame than those containing vasa 3'UTR, suggesting that nanos3 and dnd 3'UTRs are suitable for visualizing PGCs in rice field eel. Using this direct visualization method, the normal migration route of PGCs was observed from the 50%-epiboly stage to hatching stage for the first time, and the ectopic PGCs were also visualized during this period in rice field eel. These findings extend our knowledge of germ cell development, and lay a foundation for further research on the relationship between PGCs and sex differentiation, and on incubation conditions for embryos in rice field eel.
Collapse
Affiliation(s)
- Qing Xiao
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Yiqing Sun
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiao Liang
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Lihan Zhang
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Kommaly Onxayvieng
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhong Li
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China.
| | - Dapeng Li
- Hubei Provincial Engineering Laboratory for Pond Aquaculture, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
49
|
Zhang B, Zhao N, Jia L, Peng K, Che J, Li K, He X, Sun J, Bao B. Seminal Plasma Exosomes: Promising Biomarkers for Identification of Male and Pseudo-Males in Cynoglossus semilaevis. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2019; 21:310-319. [PMID: 30863906 DOI: 10.1007/s10126-019-09881-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/30/2019] [Indexed: 06/09/2023]
Abstract
In mammals, small RNAs enclosed in exosomes have been identified as appropriate signatures for disease diagnosis. However, there is limited information on exosomes derived from seminal plasma, and few studies have reported analyzed the composition of exosomes and enclosed small RNAs in fish. The half-smooth tongue sole (Cynoglossus semilaevis) is an economically important fish for aquaculture, and it exhibits sexual dimorphism: the female gender show higher growth rates and larger body sizes than males. Standard karyotype analysis and artificial gynogenesis tests have revealed that this species uses heterogametic sex determination (ZW/ZZ), and so-called sex-reversed pseudo-males exist. In this study, we successfully identified exosomes in the seminal plasma of C. semilaevis; to the best of our knowledge, this is the first report of exosomes in fish seminal plasma. Analysis of the nucleotide composition showed that miRNAs were dominant in the exosomes, and the miRNAs were sequenced and compared to identify signature miRNAs as sexual biomarkers. Moreover, target genes of the signature miRNAs were predicted by sequence matching and annotation. Finally, four miRNAs (dre-miR-141-3P, dre-miR-10d-5p, ssa-miR-27b-3p, and ssa-miR-23a-3p) with significant differential expression in the males and pseudo-males were selected from the signature candidate miRNAs as markers for sex identification, and their expression profiles were verified using real-time quantitative PCR. Our findings could provide an effective detection method for sex differentiation in fish.
Collapse
Affiliation(s)
- Bo Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University, Ministry of Education; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
- Tianjin Bohai Sea Fisheries Research Institute, Tianjin, China
| | - Na Zhao
- Tianjin Sheng Fa Biotechnology Co, Ltd, Tianjin, China
| | - Lei Jia
- Tianjin Bohai Sea Fisheries Research Institute, Tianjin, China
| | - Kang Peng
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University, Ministry of Education; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jinyuan Che
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University, Ministry of Education; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Kunming Li
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University, Ministry of Education; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiaoxu He
- Tianjin Bohai Sea Fisheries Research Institute, Tianjin, China
| | - Jinsheng Sun
- Tianjin Bohai Sea Fisheries Research Institute, Tianjin, China
| | - Baolong Bao
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University, Ministry of Education; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
50
|
Higuchi K, Goto R, Konishi J, Ina Y, Kazeto Y, Gen K. Early development of primordial germ cells in Pacific bluefin tuna Thunnus orientalis. Theriogenology 2019; 131:106-112. [DOI: 10.1016/j.theriogenology.2019.03.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/29/2019] [Accepted: 03/30/2019] [Indexed: 01/24/2023]
|