1
|
Chen Y, Ali W, Men Y, Yan K, Li Z, Cai W, He Y, Qi J. Molecular insights into oocyte development and sperm storage in black rockfish (Sebastes schlegelii): Proteomic changes across reproductive stages. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 53:101368. [PMID: 39612540 DOI: 10.1016/j.cbd.2024.101368] [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: 10/15/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/01/2024]
Abstract
Sperm storage in females is widespread among vertebrates and insects, and the expression of proteins in the female reproductive tract is influenced by the presence of sperm, allowing for adaptation to this phenomenon. Through histological observation, we confirmed that sperm were stored in the isthmic fossa outside the oocyte during the post-mating (POM) stage, and closer to the epithelial cells during the pre-fertilization (PRF) stage. In addition, we observed asynchronous ovarian development in black rockfish, where oocytes at various stages could be identified during the PRF phase. This study investigated the ovarian protein expression changes in black rockfish (Sebastes schlegelii) during key reproductive stages: pre-mating (PRM), POM, unmated control (POM-CT), and PRF. A total of 5012 proteins were identified, with notable fluctuations in protein expression observed at the PRF stage. Specifically, 140 proteins were upregulated and 615 downregulated when compared to the PRM stage, while 101 proteins were upregulated and 531 downregulated in comparison to the POM stage. The functional enrichment analysis of differentially expressed proteins (DEPs) revealed distinct pathways: POM vs. PRM showed involvement in vesicle sorting and hormone signaling; PRF vs. POM indicated pathways related to chromatin remodeling and gene expression regulation; and POM vs. POM-CT highlighted pathways associated with immune response. These findings suggested that these signaling pathways may play a crucial role in oocyte development and sperm storage. The majority of DEPs were localized in the nucleus, with key interactions involving proteins such as GSK3B and MED1. These findings enhance our understanding of the molecular mechanisms underlying oocyte maturation and sperm storage, providing insights relevant to reproductive biology and aquaculture practices.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Wajid Ali
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Yu Men
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Kai Yan
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Zibin Li
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Wenxiu Cai
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Yan He
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China
| | - Jie Qi
- Key Laboratory of Marine Genetics and Breeding, Ministry of Education/Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Qingdao 266003, Shandong, China.
| |
Collapse
|
2
|
Ocalewicz K. Quality of fish eggs and production of androgenetic and gynogenetic doubled haploids (DHs). FISH PHYSIOLOGY AND BIOCHEMISTRY 2024; 50:1947-1957. [PMID: 37296321 PMCID: PMC11576821 DOI: 10.1007/s10695-023-01206-4] [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: 12/01/2022] [Accepted: 05/19/2023] [Indexed: 06/12/2023]
Abstract
Induced development of haploid embryos (H) with only paternal (androgenesis) or maternal (gynogenesis) chromosomes requires irradiation of eggs before fertilization or activation of eggs with irradiated spermatozoa, respectively. To provide doubled haploids (DHs), androgenetic and gynogenetic haploid zygotes need to be subjected to the thermal or high hydrostatic pressure (HHP) shock to suppress the first mitotic cleavage and to double paternal or maternal haploid set of chromosomes. Androgenesis and mitotic gynogenesis (mito-gynogenesis) result in the generation of fully homozygous individuals in a single generation. DHs have been utilized in selective breeding programs, in studies concerning the phenotypic consequences of recessive alleles and to evaluate the impact of sex chromosomes on the early ontogeny. Moreover, the use of DHs for the NGS approach radically improves de novo the assembly of the genomes. However, reduced survival of the doubled haploids limits the wide application of androgenotes and gynogenotes. The high mortality of DHs may be only partly explained by the expression of recessive traits. Observed inter-clutch variation in the survival of DHs developing in eggs originating from different females make it necessary to take a closer look at the quality of the eggs used during induced androgenesis and gynogenesis. Moreover, the developmental competence of eggs that are subjected to irradiation before fertilization in order to deactivate maternal chromosomes when undergoing induced androgenesis and exposed to the physical shock after fertilization that leads to the duplication of the zygotes in both mito-gynogenesis and androgenesis may be also altered as irradiation and sublethal values of temperatures and hydrostatic pressure are considered as harmful for the cell organelles and biomolecules. Here, recently provided results concerning the morphological, biochemical, genomic, and transcriptomic characteristics of fish eggs showing high and low competence for androgenesis and mito-gynogenesis are reviewed.
Collapse
Affiliation(s)
- Konrad Ocalewicz
- Department of Marine Biology and Ecology, Institute of Oceanography, Faculty of Oceanography and Geography, University of Gdansk, Al. M. Piłsudskiego 46, 81-378, Gdynia, Poland.
| |
Collapse
|
3
|
Fuentes R, Marlow FL, Abrams EW, Zhang H, Kobayashi M, Gupta T, Kapp LD, DiNardo Z, Heller R, Cisternas R, García-Castro P, Segovia-Miranda F, Montecinos-Franjola F, Vought W, Vejnar CE, Giraldez AJ, Mullins MC. Maternal regulation of the vertebrate oocyte-to-embryo transition. PLoS Genet 2024; 20:e1011343. [PMID: 39052672 PMCID: PMC11302925 DOI: 10.1371/journal.pgen.1011343] [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: 07/09/2023] [Revised: 08/06/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
Maternally-loaded factors in the egg accumulate during oogenesis and are essential for the acquisition of oocyte and egg developmental competence to ensure the production of viable embryos. However, their molecular nature and functional importance remain poorly understood. Here, we present a collection of 9 recessive maternal-effect mutants identified in a zebrafish forward genetic screen that reveal unique molecular insights into the mechanisms controlling the vertebrate oocyte-to-embryo transition. Four genes, over easy, p33bjta, poached and black caviar, were found to control initial steps in yolk globule sizing and protein cleavage during oocyte maturation that act independently of nuclear maturation. The krang, kazukuram, p28tabj, and spotty genes play distinct roles in egg activation, including cortical granule biology, cytoplasmic segregation, the regulation of microtubule organizing center assembly and microtubule nucleation, and establishing the basic body plan. Furthermore, we cloned two of the mutant genes, identifying the over easy gene as a subunit of the Adaptor Protein complex 5, Ap5m1, which implicates it in regulating intracellular trafficking and yolk vesicle formation. The novel maternal protein Krang/Kiaa0513, highly conserved in metazoans, was discovered and linked to the function of cortical granules during egg activation. These mutant genes represent novel genetic entry points to decipher the molecular mechanisms functioning in the oocyte-to-embryo transition, fertility, and human disease. Additionally, our genetic adult screen not only contributes to the existing knowledge in the field but also sets the basis for future investigations. Thus, the identified maternal genes represent key players in the coordination and execution of events prior to fertilization.
Collapse
Affiliation(s)
- Ricardo Fuentes
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Florence L. Marlow
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine Mount Sinai, New York, New York, United States of America
| | - Elliott W. Abrams
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Biology, Purchase College, State University of New York, Purchase, New York, United States of America
| | - Hong Zhang
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Manami Kobayashi
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Tripti Gupta
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lee D. Kapp
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Zachary DiNardo
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Ronald Heller
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ruth Cisternas
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Priscila García-Castro
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Fabián Segovia-Miranda
- Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Felipe Montecinos-Franjola
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders (NIDCD), National Institutes of Health, Bethesda, Maryland, United States of America
| | - William Vought
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Charles E. Vejnar
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Antonio J. Giraldez
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Mary C. Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
4
|
Káldy J, Fazekas G, Kovács B, Molnár M, Lázár B, Pálinkás-Bodzsár N, Ljubobratović U, Fazekas G, Kovács G, Várkonyi E. Unidirectional hybridization between American paddlefish Polyodon spathula (Walbaum, 1792) and sterlet Acipenser ruthenus (Linnaeus, 1758). PeerJ 2024; 12:e16717. [PMID: 38259665 PMCID: PMC10802154 DOI: 10.7717/peerj.16717] [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: 06/23/2023] [Accepted: 12/04/2023] [Indexed: 01/24/2024] Open
Abstract
Interspecific hybridizations among sturgeon species are feasible and often bidirectional. The American paddlefish (Polyodon spathula) from Family Polyodontidae and sturgeon species from Family Acipenseridae were reported capable of hybridization, but viable hybrids have been described only in crosses with the American paddlefish as paternal parents. In the reciprocal cross, the hybrids were not viable however embryos start to develop and reach late gastrula and early neurula stages. The goal of this study was to examine the hybridization between the sterlet sturgeon (Acipenser ruthenus) and the American paddlefish. Hybrid and purebred crosses were produced by artificial fertilization. Viable hybrid offspring were harvested (three month old) and verified in the families produced by female sterlet crossing with male American paddlefish. In the reciprocal hybrid crosses with female American paddlefish and male sterlet, the embryos development did not pass over 120 h post fertilization, indicating the unidirectional hybridization between American paddlefish and sterlet. Chromosome counting showed for the same ploidy level of viable hybrid and parent species. Analysis of three microsatellite markers confirmed the unidirectional hybridization between the American paddlefish and the sterlet species. Overall, the inferred genetic cause suggests that unidirectional hybridization between American paddlefish and sterlet may be the case not only for these two species but likely also between American paddlefish and other sturgeon species.
Collapse
Affiliation(s)
- Jenő Káldy
- Research Centre for Fisheries and Aquaculture, Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, Szarvas, Békés, Hungary
| | - Georgina Fazekas
- Research Centre for Fisheries and Aquaculture, Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, Szarvas, Békés, Hungary
- PhD School of Animal Biotechnology and Animal Science, Hungarian University of Agriculture and Life Sciences, Gödöllő, Pest, Hungary
| | - Balázs Kovács
- Department of Molecular Ecology, Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, Gödöllő, Pest, Hungary
| | - Mariann Molnár
- PhD School of Animal Biotechnology and Animal Science, Hungarian University of Agriculture and Life Sciences, Gödöllő, Pest, Hungary
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Pest, Hungary
| | - Bence Lázár
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Pest, Hungary
- Animal Biotechnology Department, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, Gödöllő, Pest, Hungary
| | - Nóra Pálinkás-Bodzsár
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Pest, Hungary
| | - Uroš Ljubobratović
- Research Centre for Fisheries and Aquaculture, Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, Szarvas, Békés, Hungary
| | - Gyöngyvér Fazekas
- Research Centre for Fisheries and Aquaculture, Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, Szarvas, Békés, Hungary
| | - Gyula Kovács
- Research Centre for Fisheries and Aquaculture, Institute of Aquaculture and Environmental Safety, Hungarian University of Agriculture and Life Sciences, Szarvas, Békés, Hungary
| | - Eszter Várkonyi
- National Centre for Biodiversity and Gene Conservation, Institute for Farm Animal Gene Conservation, Gödöllő, Pest, Hungary
| |
Collapse
|
5
|
Nair S, Welch EL, Moravec CE, Trevena RL, Hansen CL, Pelegri F. The midbody component Prc1-like is required for microtubule reorganization during cytokinesis and dorsal determinant segregation in the early zebrafish embryo. Development 2023; 150:dev200564. [PMID: 36789950 PMCID: PMC10112900 DOI: 10.1242/dev.200564] [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: 08/10/2022] [Accepted: 01/10/2023] [Indexed: 02/16/2023]
Abstract
We show that the zebrafish maternal-effect mutation too much information (tmi) corresponds to zebrafish prc1-like (prc1l), which encodes a member of the MAP65/Ase1/PRC1 family of microtubule-associated proteins. Embryos from tmi homozygous mutant mothers display cytokinesis defects in meiotic and mitotic divisions in the early embryo, indicating that Prc1l has a role in midbody formation during cell division at the egg-to-embryo transition. Unexpectedly, maternal Prc1l function is also essential for the reorganization of vegetal pole microtubules required for the segregation of dorsal determinants. Whereas Prc1 is widely regarded to crosslink microtubules in an antiparallel conformation, our studies provide evidence for an additional function of Prc1l in the bundling of parallel microtubules in the vegetal cortex of the early embryo during cortical rotation and prior to mitotic cycling. These findings highlight common yet distinct aspects of microtubule reorganization that occur during the egg-to-embryo transition, driven by maternal product for the midbody component Prc1l and required for embryonic cell division and pattern formation.
Collapse
Affiliation(s)
- Sreelaja Nair
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, Maharashtra, India
| | - Elaine L. Welch
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cara E. Moravec
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ryan L. Trevena
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Christina L. Hansen
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Francisco Pelegri
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
6
|
Razmi K, Patil JG. Primordial Germ Cell Development in the Poeciliid, Gambusia holbrooki, Reveals Shared Features Between Lecithotrophs and Matrotrophs. Front Cell Dev Biol 2022; 10:793498. [PMID: 35300414 PMCID: PMC8920993 DOI: 10.3389/fcell.2022.793498] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/03/2022] [Indexed: 12/02/2022] Open
Abstract
Metazoans exhibit two modes of primordial germ cell (PGC) specification that are interspersed across taxa. However, the evolutionary link between the two modes and the reproductive strategies of lecithotrophy and matrotrophy is poorly understood. As a first step to understand this, the spatio-temporal expression of teleostean germ plasm markers was investigated in Gambusia holbrooki, a poecilid with shared lecitho- and matrotrophy. A group of germ plasm components was detected in the ovum suggesting maternal inheritance mode of PGC specification. However, the strictly zygotic activation of dnd-β and nanos1 occurred relatively early, reminiscent of models with induction mode (e.g., mice). The PGC clustering, migration and colonisation patterns of G. holbrooki resembled those of zebrafish, medaka and mice at blastula, gastrula and somitogenesis, respectively-recapitulating features of advancing evolutionary nodes with progressive developmental stages. Moreover, the expression domains of PGC markers in G. holbrooki were either specific to teleost (vasa expression in developing PGCs), murine models (dnd spliced variants) or shared between the two taxa (germline and somatic expression of piwi and nanos1). Collectively, the results suggest that the reproductive developmental adaptations may reflect a transition from lecithotrophy to matrotrophy.
Collapse
Affiliation(s)
- Komeil Razmi
- Laboratory of Molecular Biology, Fisheries and Aquaculture Centre, Institute for Marine and Antarctic Studies, University of Tasmania, Taroona, TAS, Australia
| | - Jawahar G. Patil
- Laboratory of Molecular Biology, Fisheries and Aquaculture Centre, Institute for Marine and Antarctic Studies, University of Tasmania, Taroona, TAS, Australia
| |
Collapse
|
7
|
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
|
8
|
Zhang C, Li J, Tarique I, Zhang Y, Lu T, Wang J, Chen A, Wen F, Zhang Z, Zhang Y, Shao M. A Time-Saving Strategy to Generate Double Maternal Mutants by an Oocyte-Specific Conditional Knockout System in Zebrafish. BIOLOGY 2021; 10:biology10080777. [PMID: 34440009 PMCID: PMC8389640 DOI: 10.3390/biology10080777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/29/2021] [Accepted: 08/14/2021] [Indexed: 12/23/2022]
Abstract
Simple Summary Maternally supplied mRNAs and proteins, termed maternal factors, are produced by over 14,000 coding genes in zebrafish. They play exclusive roles in controlling the formation of oocytes and the development of early embryos. These maternal factors can also compensate for the loss of function of its corresponding zygotic gene products. Thus, eliminating both maternal and zygotic gene products is essential to elucidate the functions of more than half of zebrafish genes. However, it is always challenging to inactivate maternal factors, because traditional genetic methods are either technically demanding or time-consuming. Our recent work established a rapid conditional knockout method to generate maternal or maternal and zygotic mutants in one fish generation. Here, we further test the feasibility of this approach to knock out two maternal genes with functional redundancy simultaneously. As a proof of principle, we successfully generated double maternal mutant embryos for dvl2 and dvl3a genes in three months for the first time. The cell movement defects in mutant embryos obtained by this approach mimic the genuine mutant embryos generated after fifteen months of time-consuming screening following the previously reported mosaic strategy. Therefore, this method has the potential to speed up the functional study of paralogous maternal genes. Abstract Maternal products are those mRNAs and proteins deposited during oogenesis, which play critical roles in controlling oocyte formation, fertilization, and early embryonic development. However, loss-of-function studies for these maternal factors are still lacking, mainly because of the prolonged period of transgenerational screening and technical barriers that prevent the generation of maternal (M) and maternal and zygotic (MZ) mutant embryos. By the transgenic expression of multiple sgRNAs targeting a single gene of interest in the background of a transgenic line Tg(zpc:zcas9) with oocyte-specific cas9 expression, we have successfully obtained maternal or maternal–zygotic mutant for single genes in F1 embryos. In this work, we tandemly connected a maternal GFP marker and eight sgRNA expression units to target dvl2 and dvl3a simultaneously and introduced this construct to the genome of Tg(zpc:zcas9) by meganuclease I-Sce I. As expected, we confirmed the existence of Mdvl2;Mdvl3a embryos with strong defective convergence and extension movement during gastrulation among outcrossed GFP positive F1 offspring. The MZdvl2;MZdvl3a embryos were also obtained by crossing the mutant carrying mosaic F0 female with dvl2+/−;dvl3a−/− male fish. This proof-of-principle thus highlights the potential of this conditional knockout strategy to circumvent the current difficulty in the study of genes with multiple functionally redundant paralogs.
Collapse
Affiliation(s)
- Chong Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Jiaguang Li
- Taishan College, Shandong University, Qingdao 266237, China; (J.L.); (Z.Z.)
| | - Imran Tarique
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Yizhuang Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Tong Lu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Jiasheng Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Aijun Chen
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Fenfen Wen
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Zhuoyu Zhang
- Taishan College, Shandong University, Qingdao 266237, China; (J.L.); (Z.Z.)
| | - Yanjun Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
| | - Ming Shao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China; (C.Z.); (I.T.); (Y.Z.); (T.L.); (J.W.); (A.C.); (F.W.); (Y.Z.)
- Taishan College, Shandong University, Qingdao 266237, China; (J.L.); (Z.Z.)
- Correspondence:
| |
Collapse
|
9
|
Zhang C, Lu T, Zhang Y, Li J, Tarique I, Wen F, Chen A, Wang J, Zhang Z, Zhang Y, Shi DL, Shao M. Rapid generation of maternal mutants via oocyte transgenic expression of CRISPR-Cas9 and sgRNAs in zebrafish. SCIENCE ADVANCES 2021; 7:eabg4243. [PMID: 34362733 PMCID: PMC8346210 DOI: 10.1126/sciadv.abg4243] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/21/2021] [Indexed: 05/08/2023]
Abstract
Maternal products are exclusive factors to drive oogenesis and early embryonic development. As disrupting maternal gene functions is either time-consuming or technically challenging, early developmental programs regulated by maternal factors remain mostly elusive. We provide a transgenic approach to inactivate maternal genes in zebrafish primary oocytes. By introducing three tandem single guide RNA (sgRNA) expression cassettes and a green fluorescent protein (GFP) reporter into Tg(zpc:zcas9) embryos, we efficiently obtained maternal nanog and ctnnb2 mutants among GFP-positive F1 offspring. Notably, most of these maternal mutants displayed either sgRNA site-spanning genomic deletions or unintended large deletions extending distantly from the sgRNA targets, suggesting a prominent deletion-prone tendency of genome editing in the oocyte. Thus, our method allows maternal gene knockout in the absence of viable and fertile homozygous mutant adults. This approach is particularly time-saving and can be applied for functional screening of maternal factors and generating genomic deletions in zebrafish.
Collapse
Affiliation(s)
- Chong Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Tong Lu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Yizhuang Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Jiaguang Li
- Shandong University Taishan College, Qingdao 266237, China
| | - Imran Tarique
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Fenfen Wen
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Aijun Chen
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Jiasheng Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Zhuoyu Zhang
- Shandong University Taishan College, Qingdao 266237, China
| | - Yanjun Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China
| | - De-Li Shi
- Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
- Developmental Biology Laboratory, CNRS-UMR7622, Institut de Biologie Paris-Seine, Sorbonne University, Paris 75005, France
| | - Ming Shao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology and Key Laboratory for Experimental Teratology of the Ministry of Education, School of Life Sciences, Shandong University, Qingdao 266237, China.
- Shandong University Taishan College, Qingdao 266237, China
| |
Collapse
|
10
|
Crichigno SA, Orellana M, Larraza R, Mirenna G, Cussac VE. Thermal effects in rainbow trout (Oncorhynchus mykiss) F1 embryos (farmed female × wild thermal-resistant male). JOURNAL OF FISH BIOLOGY 2021; 99:197-205. [PMID: 33625760 DOI: 10.1111/jfb.14711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 06/12/2023]
Abstract
The aim of this work was to investigate the response of rainbow trout embryos (Oncorhynchus mykiss) (i.e., survival, size at hatching, time to hatching, malformations) to four incubation temperatures (5.8, 8.9, 14.0 and 16.8°C), taking into account the origin of the male parental genome and comparing pure farmed and F1 embryos (farmed female × wild thermal-resistant male). Several consequences of thermal stress were observed: lower accumulated thermal units (ATU) at hatching at high temperatures, and lower survival, shorter hatched free embryos and less-consumed yolk sac at extreme temperatures. The effect of the thermal-adapted male parental genome was shown only in the lower percentage of incompletely hatched free embryos in the F1 families. It appears that to obtain greater modification of thermal performance during early development, the adapted genome of the wild thermal-resistant population has to be included through maternal inheritance, thus producing a stabilized strain selected for domesticity, growth and thermal adaptation.
Collapse
Affiliation(s)
- Sonia A Crichigno
- Instituto Patagónico de Tecnologías Biológicas y Geoambientales, Universidad Nacional del Comahue - Consejo Nacional de Investigaciones Científicas y Técnicas, Bariloche, Argentina
| | - Mabel Orellana
- Centro de Salmonicultura Bariloche, Universidad Nacional del Comahue, Neuquen, Argentina
| | - Rodrigo Larraza
- Centro de Salmonicultura Bariloche, Universidad Nacional del Comahue, Neuquen, Argentina
| | - Guillermo Mirenna
- Centro de Salmonicultura Bariloche, Universidad Nacional del Comahue, Neuquen, Argentina
| | - Víctor E Cussac
- Instituto Patagónico de Tecnologías Biológicas y Geoambientales, Universidad Nacional del Comahue - Consejo Nacional de Investigaciones Científicas y Técnicas, Bariloche, Argentina
| |
Collapse
|
11
|
Applying Rho Pathway Inhibitors to Investigate Germ Plasm Localization. Methods Mol Biol 2021. [PMID: 33606225 DOI: 10.1007/978-1-0716-0970-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
The correct assembly, migration, and segregation of the mRNAs of the germ plasm during the first cell divisions are intimately connected to the cytoskeleton and cytokinesis.RhoA is a key regulator of germ plasm localization during the first two cell division cycles in zebrafish embryos. Pharmacological inhibition of RhoA and his effector ROCK affected the correct assembly of microtubules in the cleavage furrow with the concomitant abnormal localization of germ plasm mRNAs. The inhibition of RhoA/ROCK pathway caused a significant decrease in the germ cell population later in development.
Collapse
|
12
|
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
|
13
|
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
|
14
|
Romney ALT, Yanagitsuru YR, Mundy PC, Fangue NA, Hung TC, Brander SM, Connon RE. Developmental Staging and Salinity Tolerance in Embryos of the Delta Smelt, Hypomesus transpacificus. AQUACULTURE (AMSTERDAM, NETHERLANDS) 2019; 511:634191. [PMID: 32831418 PMCID: PMC7442155 DOI: 10.1016/j.aquaculture.2019.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Delta smelt (Hypomesus transpacificus) is a critically endangered species endemic to the San Francisco Bay Delta (SFBD). Important for the conservation of this species is understanding the physiological and ecological impacts contributing to their population decline, and current studies lack information on embryonic development. Changes in patterns of salinity across the SFBD may be a particularly important environmental stressor contributing to the recruitment and survival of the species. Throughout their ontogeny, delta smelt may exhibit unique requirements and tolerances to environmental conditions including salinity. Here, we describe 22 stages of embryonic development of H. transpacificus that characterize early differentiation from the fertilized egg until hatching, allowing the identification of critical morphological features unique to this species. Additionally, we investigated aspects of physiological tolerance to environmental salinity during development. Embryos survived incubation at salinity treatments between 0.4 and 20 ppt, yet had lower hatch success at higher salinities. Prior to hatching, embryos exposed to higher salinities had increased osmolalities and reduced fractions of yolk implying that the elevated external salinity altered the physiology of the embryo and the environment internal to the chorion. Lastly, egg activation and fertilization appear to also be impacted by salinity. Altogether, we suggest that any potential tolerance to salinity during embryogenesis, a common feature in euryhaline teleost species, impacts life cycle transitions into, and out of, embryonic development. Results from this investigation should improve conservation and management practices of this species and further expand our understanding of the intimate relationship between an embryo and its environment.
Collapse
Affiliation(s)
- Amie L. T. Romney
- Department of Anatomy, Physiology, and Cell Biology, University of California, Davis, CA 95616, USA
| | - Yuzo R. Yanagitsuru
- Department of Wildlife Conservation and Fish Biology, University of California, Davis, CA 95616, USA
| | - Paige C. Mundy
- Department of Anatomy, Physiology, and Cell Biology, University of California, Davis, CA 95616, USA
| | - Nann A. Fangue
- Department of Wildlife Conservation and Fish Biology, University of California, Davis, CA 95616, USA
| | - Tien-Chieh Hung
- Fish Conservation Culture Laboratory, Department of Biological and Agricultural Engineering, University of California, Davis, CA 95616, USA
| | - Susanne M. Brander
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Richard E. Connon
- Department of Anatomy, Physiology, and Cell Biology, University of California, Davis, CA 95616, USA
| |
Collapse
|
15
|
Fuentes R, Letelier J, Tajer B, Valdivia LE, Mullins MC. Fishing forward and reverse: Advances in zebrafish phenomics. Mech Dev 2018; 154:296-308. [PMID: 30130581 PMCID: PMC6289646 DOI: 10.1016/j.mod.2018.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 08/06/2018] [Accepted: 08/17/2018] [Indexed: 12/15/2022]
Abstract
Understanding how the genome instructs the phenotypic characteristics of an organism is one of the major scientific endeavors of our time. Advances in genetics have progressively deciphered the inheritance, identity and biological relevance of genetically encoded information, contributing to the rise of several, complementary omic disciplines. One of them is phenomics, an emergent area of biology dedicated to the systematic multi-scale analysis of phenotypic traits. This discipline provides valuable gene function information to the rapidly evolving field of genetics. Current molecular tools enable genome-wide analyses that link gene sequence to function in multi-cellular organisms, illuminating the genome-phenome relationship. Among vertebrates, zebrafish has emerged as an outstanding model organism for high-throughput phenotyping and modeling of human disorders. Advances in both systematic mutagenesis and phenotypic analyses of embryonic and post-embryonic stages in zebrafish have revealed the function of a valuable collection of genes and the general structure of several complex traits. In this review, we summarize multiple large-scale genetic efforts addressing parental, embryonic, and adult phenotyping in the zebrafish. The genetic and quantitative tools available in the zebrafish model, coupled with the broad spectrum of phenotypes that can be assayed, make it a powerful model for phenomics, well suited for the dissection of genotype-phenotype associations in development, physiology, health and disease.
Collapse
Affiliation(s)
- Ricardo Fuentes
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joaquín Letelier
- Centro Andaluz de Biología del Desarrollo (CSIC/UPO/JA), Seville, Spain; Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Benjamin Tajer
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leonardo E Valdivia
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago, Chile.
| | - Mary C Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Cheung CT, Pasquier J, Bouleau A, Nguyen T, Chesnel F, Guiguen Y, Bobe J. Double maternal-effect: duplicated nucleoplasmin 2 genes, npm2a and npm2b, with essential but distinct functions are shared by fish and tetrapods. BMC Evol Biol 2018; 18:167. [PMID: 30419815 PMCID: PMC6233590 DOI: 10.1186/s12862-018-1281-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 10/26/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Nucleoplasmin 2 (npm2) is an essential maternal-effect gene that mediates early embryonic events through its function as a histone chaperone that remodels chromatin. Recently, two npm2 (npm2a and npm2b) genes have been annotated in zebrafish. Thus, we examined the evolution of npm2a and npm2b in a variety of vertebrates, their potential phylogenetic relationships, and their biological functions using knockout models via the CRISPR/cas9 system. RESULTS We demonstrated that the two npm2 duplicates exist in a wide range of vertebrates, including sharks, ray-finned fish, amphibians, and sauropsids, while npm2a was lost in coelacanth and mammals, as well as some specific teleost lineages. Using phylogeny and synteny analyses, we traced their origins to the early stages of vertebrate evolution. Our findings suggested that npm2a and npm2b resulted from an ancient local gene duplication, and their functions diverged although key protein domains were conserved. We then investigated their functions by examining their tissue distribution in a wide variety of species and found that they shared ovarian-specific expression, a key feature of maternal-effect genes. We also demonstrated that both npm2a and npm2b are maternally-inherited transcripts in vertebrates, and that they play essential, but distinct, roles in early embryogenesis using zebrafish knockout models. Both npm2a and npm2b function early during oogenesis and may play a role in cortical granule function that impact egg activation and fertilization, while npm2b is also involved in early embryogenesis. CONCLUSION These novel findings will broaden our knowledge on the evolutionary history of maternal-effect genes and underlying mechanisms that contribute to vertebrate reproductive success. In addition, our results demonstrate the existence of a newly described maternal-effect gene, npm2a, that contributes to egg competence, an area that still requires further comprehension.
Collapse
Affiliation(s)
| | | | | | - Thaovi Nguyen
- INRA LPGP UR1037, Campus de Beaulieu, 35042, Rennes, France
| | - Franck Chesnel
- CNRS/UMR6290, Université de Rennes 1, 35000, Rennes, France
| | - Yann Guiguen
- INRA LPGP UR1037, Campus de Beaulieu, 35042, Rennes, France
| | - Julien Bobe
- INRA LPGP UR1037, Campus de Beaulieu, 35042, Rennes, France. .,Laboratory of fish physiology and genomics (LPGP), National Institute of Agricultural Research (INRA), Campus de Beaulieu, 35042, Rennes Cedex, France.
| |
Collapse
|
17
|
Romney ALT, Podrabsky JE. Small noncoding RNA profiles along alternative developmental trajectories in an annual killifish. Sci Rep 2018; 8:13364. [PMID: 30190591 PMCID: PMC6127099 DOI: 10.1038/s41598-018-31466-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 08/13/2018] [Indexed: 11/24/2022] Open
Abstract
Embryonic development of Austrofundulus limnaeus can occur along two phenotypic trajectories that are physiologically and biochemically distinct. Phenotype appears to be influenced by maternal provisioning based on the observation that young females produce predominately non-diapausing embryos and older females produce mostly diapausing embryos. Embryonic incubation temperature can override this pattern and alter trajectory. We hypothesized that temperature-induced phenotypic plasticity may be regulated by post-transcriptional modification via noncoding RNAs. As a first step to exploring this possibility, RNA-seq was used to generate transcriptomic profiles of small noncoding RNAs in embryos developing along the two alternative trajectories. We find distinct profiles of mature sequences belonging to the miR-10 family expressed in increasing abundance during development and mature sequences of miR-430 that follow the opposite pattern. Furthermore, miR-430 sequences are enriched in escape trajectory embryos. MiR-430 family members are known to target maternally provisioned mRNAs in zebrafish and may operate similarly in A. limnaeus in the context of normal development, and also by targeting trajectory-specific mRNAs. This expression pattern and function for miR-430 presents a potentially novel model for maternal-embryonic conflict in gene regulation that provides the embryo the ability to override maternal programming in the face of altered environmental conditions.
Collapse
Affiliation(s)
- Amie L T Romney
- Department of Biology, Portland State University, P.O. Box 751, Portland, OR, 97207, USA.
- Department of Anatomy, Physiology & Cell Biology, University of California at Davis School of Veterinary Medicine, One Shields Ave, Davis, CA, 95616, USA.
| | - Jason E Podrabsky
- Department of Biology, Portland State University, P.O. Box 751, Portland, OR, 97207, USA.
| |
Collapse
|
18
|
Cheung CT, Patinote A, Guiguen Y, Bobe J. foxr1 is a novel maternal-effect gene in fish that is required for early embryonic success. PeerJ 2018; 6:e5534. [PMID: 30155373 PMCID: PMC6109588 DOI: 10.7717/peerj.5534] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/08/2018] [Indexed: 01/16/2023] Open
Abstract
The family of forkhead box (Fox) transcription factors regulates gonadogenesis and embryogenesis, but the role of foxr1 in reproduction is unknown. Evolutionary history of foxr1 in vertebrates was examined and the gene was found to exist in most vertebrates, including mammals, ray-finned fish, amphibians, and sauropsids. By quantitative PCR and RNA-seq, we found that foxr1 had an ovarian-specific expression in zebrafish, a common feature of maternal-effect genes. In addition, it was demonstrated using in situ hybridization that foxr1 was a maternally-inherited transcript that was highly expressed even in early-stage oocytes and accumulated in the developing eggs during oogenesis. We also analyzed the function of foxr1 in female reproduction using a zebrafish CRISPR/cas9 knockout model. It was observed that embryos from the foxr1-deficient females had a significantly lower survival rate whereby they either failed to undergo cell division or underwent abnormal division that culminated in growth arrest at around the mid-blastula transition and early death. These mutant-derived eggs contained dramatically increased levels of p21, a cell cycle inhibitor, and reduced rictor, a component of mTOR and regulator of cell survival, which were in line with the observed growth arrest phenotype. Our study shows for the first time that foxr1 is an essential maternal-effect gene and may be required for proper cell division and survival via the p21 and mTOR pathways. These novel findings will broaden our knowledge on the functions of specific maternal factors stored in the developing egg and the underlying mechanisms that contribute to reproductive success.
Collapse
Affiliation(s)
| | - Amélie Patinote
- LPGP, UR1037 Fish Physiology and Genomics, INRA, Rennes, France
| | - Yann Guiguen
- LPGP, UR1037 Fish Physiology and Genomics, INRA, Rennes, France
| | - Julien Bobe
- LPGP, UR1037 Fish Physiology and Genomics, INRA, Rennes, France
| |
Collapse
|
19
|
Fuentes R, Mullins MC, Fernández J. Formation and dynamics of cytoplasmic domains and their genetic regulation during the zebrafish oocyte-to-embryo transition. Mech Dev 2018; 154:259-269. [PMID: 30077623 DOI: 10.1016/j.mod.2018.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022]
Abstract
Establishment and movement of cytoplasmic domains is of great importance for the emergence of cell polarity, germline segregation, embryonic axis specification and correct sorting of organelles and macromolecules into different embryonic cells. The zebrafish oocyte, egg and zygote are valuable material for the study of cytoplasmic domains formation and dynamics during development. In this review we examined how cytoplasmic domains form and are relocated during zebrafish early embryogenesis. Distinct cortical cytoplasmic domains (also referred to as ectoplasm domains) form first during early oogenesis by the localization of mRNAs to the vegetal or animal poles of the oocyte or radially throughout the cortex. Cytoplasmic segregation in the late oocyte relocates non-cortical cytoplasm (endoplasm) into the preblastodisc and yolk cell. The preblastodisc is a precursor to the blastodisc, which gives rise to the blastoderm and most the future embryo. After egg activation, the blastodisc enlarges by transport of cytoplasm from the yolk cell to the animal pole, along defined pathways or streamers that include a complex cytoskeletal meshwork and cytoplasmic movement at different speeds. A powerful actin ring, assembled at the margin of the blastodisc, appears to drive the massive streaming of cytoplasm. The fact that the mechanism(s) leading to the formation and relocation of cytoplasmic domains are affected in maternal-effect mutants indicates that these processes are under maternal control. Here, we also discuss why these mutants represent outstanding genetic entry points to investigate the genetic basis of cytoplasmic segregation. Functional studies, combined with the analysis of zebrafish mutants, generated by forward and reverse genetic strategies, are expected to decipher the molecular mechanism(s) by which the maternal factors regulate cytoplasmic movements during early vertebrate development.
Collapse
Affiliation(s)
- Ricardo Fuentes
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Juan Fernández
- Department of Biology, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
20
|
Welch EL, Eno CC, Nair S, Lindeman RE, Pelegri F. Functional Manipulation of Maternal Gene Products Using In Vitro Oocyte Maturation in Zebrafish. J Vis Exp 2017. [PMID: 28518112 PMCID: PMC5565100 DOI: 10.3791/55213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cellular events that take place during the earliest stages of animal embryonic development are driven by maternally derived gene products deposited into the developing oocyte. Because these events rely on maternal products which typically act very soon after fertilization-that preexist inside the egg, standard approaches for expression and functional reduction involving the injection of reagents into the fertilized egg are typically ineffective. Instead, such manipulations must be performed during oogenesis, prior to or during the accumulation of maternal products. This article describes in detail a protocol for the in vitro maturation of immature zebrafish oocytes and their subsequent in vitro fertilization, yielding viable embryos that survive to adulthood. This method allows the functional manipulation of maternal products during oogenesis, such as the expression of products for phenotypic rescue and tagged construct visualization, as well as the reduction of gene function through reverse-genetics agents.
Collapse
Affiliation(s)
| | - Celeste C Eno
- Laboratory of Genetics, University of Wisconsin-Madison
| | - Sreelaja Nair
- Department of Biological Sciences, Tata Institute of Fundamental Research
| | - Robin E Lindeman
- Department of Genetics, Cell Biology, and Development, University of Minnesota
| | | |
Collapse
|
21
|
DND protein functions as a translation repressor during zebrafish embryogenesis. Biochem Biophys Res Commun 2017; 484:235-240. [PMID: 28115159 DOI: 10.1016/j.bbrc.2017.01.080] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/17/2017] [Indexed: 11/23/2022]
Abstract
Germline and somatic cell distinction is regulated through a combination of microRNA and germ cell-specific RNA-binding proteins in zebrafish. An RNA-binding protein, DND, has been reported to relieve the miR-430-mediated repression of some germ plasm mRNAs such as nanos3 and tdrd7 in primordial germ cells (PGCs). Here, we showed that miR-430-mediated repression is not counteracted by the overexpression of DND protein in somatic cells. Using a λN-box B tethering assay in the embryo, we found that tethering of DND to reporter mRNA results in translation repression without affecting mRNA stability. Translation repression by DND was not dependent on another germline-specific translation repressor, Nanos3, in zebrafish embryos. Moreover, our data suggested that DND represses translation of nanog and dnd mRNAs, whereas an RNA-binding protein DAZ-like (DAZL) promotes dnd mRNA translation. Thus, our study showed that DND protein functions as a translation repressor of specific mRNAs to control PGC development in zebrafish.
Collapse
|
22
|
Miranda-Rodríguez JR, Salas-Vidal E, Lomelí H, Zurita M, Schnabel D. RhoA/ROCK pathway activity is essential for the correct localization of the germ plasm mRNAs in zebrafish embryos. Dev Biol 2016; 421:27-42. [PMID: 27836552 DOI: 10.1016/j.ydbio.2016.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 10/01/2016] [Accepted: 11/01/2016] [Indexed: 01/01/2023]
Abstract
Zebrafish germ plasm is composed of mRNAs such as vasa and nanos and of proteins such as Bucky ball, all of which localize symmetrically in four aggregates at the distal region of the first two cleavage furrows. The coordination of actin microfilaments, microtubules and kinesin is essential for the correct localization of the germ plasm. Rho-GTPases, through their effectors, coordinate cytoskeletal dynamics. We address the participation of RhoA and its effector ROCK in germ plasm localization during the transition from two- to eight-cell embryos. We found that active RhoA is enriched along the cleavage furrow during the first two division cycles, whereas ROCK localizes at the distal region of the cleavage furrows in a similar pattern as the germ plasm mRNAs. Specific inhibition of RhoA and ROCK affected microtubules organization at the cleavage furrow; these caused the incorrect localization of the germ plasm mRNAs. The incorrect localization of the germ plasm led to a dramatic change in the number of germ cells during the blastula and 24hpf embryo stages without affecting any other developmental processes. We demonstrate that the Rho/ROCK pathway is intimately related to the determination of germ cells in zebrafish embryos.
Collapse
Affiliation(s)
- Jerónimo Roberto Miranda-Rodríguez
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, AP 510-3, Cuernavaca, Mor. 62250, Mexico
| | - Enrique Salas-Vidal
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, AP 510-3, Cuernavaca, Mor. 62250, Mexico
| | - Hilda Lomelí
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, AP 510-3, Cuernavaca, Mor. 62250, Mexico
| | - Mario Zurita
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, AP 510-3, Cuernavaca, Mor. 62250, Mexico
| | - Denhi Schnabel
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, AP 510-3, Cuernavaca, Mor. 62250, Mexico.
| |
Collapse
|
23
|
Lombard-Banek C, Moody SA, Nemes P. High-Sensitivity Mass Spectrometry for Probing Gene Translation in Single Embryonic Cells in the Early Frog ( Xenopus) Embryo. Front Cell Dev Biol 2016; 4:100. [PMID: 27761436 PMCID: PMC5050209 DOI: 10.3389/fcell.2016.00100] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/29/2016] [Indexed: 01/01/2023] Open
Abstract
Direct measurement of protein expression with single-cell resolution promises to deepen the understanding of the basic molecular processes during normal and impaired development. High-resolution mass spectrometry provides detailed coverage of the proteomic composition of large numbers of cells. Here we discuss recent mass spectrometry developments based on single-cell capillary electrophoresis that extend discovery proteomics to sufficient sensitivity to enable the measurement of proteins in single cells. The single-cell mass spectrometry system is used to detect a large number of proteins in single embryonic cells in the 16-cell embryo of the South African clawed frog (Xenopus laevis) that give rise to distinct tissue types. Single-cell measurements of protein expression provide complementary information on gene transcription during early development of the vertebrate embryo, raising a potential to understand how differential gene expression coordinates normal cell heterogeneity during development.
Collapse
Affiliation(s)
| | - Sally A Moody
- Department of Anatomy and Regenerative Biology, The George Washington University Washington, DC, USA
| | - Peter Nemes
- Department of Chemistry, The George Washington University Washington, DC, USA
| |
Collapse
|
24
|
Pelegri F, Mullins MC. Genetic screens for mutations affecting adult traits and parental-effect genes. Methods Cell Biol 2016; 135:39-87. [PMID: 27443920 DOI: 10.1016/bs.mcb.2016.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Forward genetics remains an important approach for the unbiased identification of factors involved in biological pathways. Forward genetic analysis in the zebrafish has until now largely been restricted to the developmental period from zygotic genome activation through the end of embryogenesis. However, the use of the zebrafish as a model system for the analysis of late larval, juvenile and adult traits, including fertility and maternal and paternal effects, continues to gain momentum. Here, we describe two approaches, based on an F3-extended family and gynogenetic methods, that allow genetic screening for, and recovery of mutations affecting post-embryonic stages, including adult traits, fertility, and parental effects. For each approach, we also describe strategies to maintain, map, and molecularly clone the identified mutations.
Collapse
Affiliation(s)
- F Pelegri
- University of Wisconsin-Madison, Madison, WI, United States
| | - M C Mullins
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
25
|
Rauwerda H, Wackers P, Pagano JFB, de Jong M, Ensink W, Dekker R, Nehrdich U, Spaink HP, Jonker M, Breit TM. Mother-Specific Signature in the Maternal Transcriptome Composition of Mature, Unfertilized Zebrafish Eggs. PLoS One 2016; 11:e0147151. [PMID: 26799215 PMCID: PMC4723340 DOI: 10.1371/journal.pone.0147151] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 12/28/2015] [Indexed: 12/19/2022] Open
Abstract
Maternal mRNA present in mature oocytes plays an important role in the proper development of the early embryo. As the composition of the maternal transcriptome in general has been studied with pooled mature eggs, potential differences between individual eggs are unknown. Here we present a transcriptome study on individual zebrafish eggs from clutches of five mothers in which we focus on the differences in maternal mRNA abundance per gene between and within clutches. To minimize technical interference, we used mature, unfertilized eggs from siblings. About half of the number of analyzed genes was found to be expressed as maternal RNA. The expressed and non-expressed genes showed that maternal mRNA accumulation is a non-random process, as it is related to specific biological pathways and processes relevant in early embryogenesis. Moreover, it turned out that overall the composition of the maternal transcriptome is tightly regulated as about half of the expressed genes display a less than twofold expression range between the observed minimum and maximum expression values of a gene in the experiment. Even more, the maximum gene-expression difference within clutches is for 88% of the expressed genes lower than twofold. This means that expression differences observed in maternally expressed genes are primarily caused by differences between mothers, with only limited variability between eggs from the same mother. This was underlined by the fact that 99% of the expressed genes were found to be differentially expressed between any of the mothers in an ANOVA test. Furthermore, linking chromosome location, transcription factor binding sites, and miRNA target sites of the genes in clusters of distinct and unique mother-specific gene-expression, suggest biological relevance of the mother-specific signatures in the maternal transcriptome composition. Altogether, the maternal transcriptome composition of mature zebrafish oocytes seems to be tightly regulated with a distinct mother-specific signature.
Collapse
Affiliation(s)
- Han Rauwerda
- RNA Biology & Applied Bioinformatics research group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Paul Wackers
- RNA Biology & Applied Bioinformatics research group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Johanna F. B. Pagano
- RNA Biology & Applied Bioinformatics research group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Mark de Jong
- RNA Biology & Applied Bioinformatics research group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Wim Ensink
- RNA Biology & Applied Bioinformatics research group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Rob Dekker
- RNA Biology & Applied Bioinformatics research group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Ulrike Nehrdich
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, the Netherlands
| | - Herman P. Spaink
- Institute of Biology Leiden, Faculty of Science, Leiden University, Leiden, the Netherlands
| | - Martijs Jonker
- RNA Biology & Applied Bioinformatics research group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| | - Timo M. Breit
- RNA Biology & Applied Bioinformatics research group, Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Elkouby YM, Jamieson-Lucy A, Mullins MC. Oocyte Polarization Is Coupled to the Chromosomal Bouquet, a Conserved Polarized Nuclear Configuration in Meiosis. PLoS Biol 2016; 14:e1002335. [PMID: 26741740 PMCID: PMC4704784 DOI: 10.1371/journal.pbio.1002335] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 11/19/2015] [Indexed: 12/02/2022] Open
Abstract
The source of symmetry breaking in vertebrate oocytes is unknown. Animal—vegetal oocyte polarity is established by the Balbiani body (Bb), a conserved structure found in all animals examined that contains an aggregate of specific mRNAs, proteins, and organelles. The Bb specifies the oocyte vegetal pole, which is key to forming the embryonic body axes as well as the germline in most vertebrates. How Bb formation is regulated and how its asymmetric position is established are unknown. Using quantitative image analysis, we trace oocyte symmetry breaking in zebrafish to a nuclear asymmetry at the onset of meiosis called the chromosomal bouquet. The bouquet is a universal feature of meiosis where all telomeres cluster to one pole on the nuclear envelope, facilitating chromosomal pairing and meiotic recombination. We show that Bb precursor components first localize with the centrosome to the cytoplasm adjacent to the telomere cluster of the bouquet. They then aggregate around the centrosome in a specialized nuclear cleft that we identified, assembling the early Bb. We show that the bouquet nuclear events and the cytoplasmic Bb precursor localization are mechanistically coordinated by microtubules. Thus the animal—vegetal axis of the oocyte is aligned to the nuclear axis of the bouquet. We further show that the symmetry breaking events lay upstream to the only known regulator of Bb formation, the Bucky ball protein. Our findings link two universal features of oogenesis, the Bb and the chromosomal bouquet, to oocyte polarization. We propose that a meiotic—vegetal center couples meiosis and oocyte patterning. Our findings reveal a novel mode of cellular polarization in meiotic cells whereby cellular and nuclear polarity are aligned. We further reveal that in zygotene nests, intercellular cytoplasmic bridges remain between oocytes and that the position of the cytoplasmic bridge coincides with the location of the centrosome meiotic—vegetal organizing center. These results suggest that centrosome positioning is set by the last mitotic oogonial division plane. Thus, oocytes are polarized in two steps: first, mitotic divisions preset the centrosome with no obvious polarization yet, then the meiotic—vegetal center forms at zygotene bouquet stages, when symmetry is, in effect, broken. This study traces symmetry breaking in zebrafish oocytes to a cellular organizer that controls the configuration of the meiotic polarized chromosomal bouquet, thereby coupling meiosis and oocyte patterning at the nexus of oocyte differentiation. In most vertebrates, an early event in egg development involves the establishment of the so-called animal—vegetal axis; this sets up the embryonic body axes and contributes to germ-line specification, and therefore, is key to embryonic development. The animal—vegetal axis is established during oogenesis by the Balbiani body (Bb), an aggregate of specific mRNAs, proteins, and mitochondria, which forms adjacent to the nucleus and ultimately defines one pole of the oocyte, the vegetal pole. Despite its universal conservation, how the Bb forms and how its position is determined is unknown. Here, we show that Bb formation is initiated at the onset of meiosis, and its position coincides with a previously known meiotic polarized nuclear configuration, the chromosomal bouquet, which gathers the chromosome ends, the telomeres, asymmetrically on the nuclear membrane to assist in homologous chromosome pairing. We reveal that a global cellular organizer functioning via microtubules generates the bouquet and aggregates the Bb precursors asymmetrically towards the centrosome. We determined that these events lie functionally upstream to the Bb regulator Bucky ball. Further upstream, we found that the centrosome appears prepositioned by an intercellular cytoplasmic bridge derived from the last presumptive cell division plane of the premeiotic oogonial cell. Thus, oocyte polarity and the chromosomal bouquet are linked through a common cellular polarization mechanism.
Collapse
Affiliation(s)
- Yaniv M. Elkouby
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Allison Jamieson-Lucy
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mary C. Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
27
|
Riemer S, Bontems F, Krishnakumar P, Gömann J, Dosch R. A functional Bucky ball-GFP transgene visualizes germ plasm in living zebrafish. Gene Expr Patterns 2015; 18:44-52. [PMID: 26143227 DOI: 10.1016/j.gep.2015.05.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 05/08/2015] [Accepted: 05/09/2015] [Indexed: 10/23/2022]
Abstract
In many animals, the germline is specified by maternal RNA-granules termed germ plasm. The correct localization of germ plasm during embryogenesis is therefore crucial for the specification of germ cells. In zebrafish, we previously identified Bucky ball (Buc) as a key regulator of germ plasm formation. Here, we used a Buc antibody to describe its continuous germ plasm localization. Moreover, we generated a transgenic Buc-GFP line for live imaging, which visualizes germ plasm from its assembly during oogenesis up to the larval stages. Live imaging of Buc-GFP generated stunning movies, as they highlighted the dynamic details of germ plasm movements. Moreover, we discovered that Buc was still detected in primordial germ cells 2 days after fertilization. Interestingly, the transgene rescued buc mutants demonstrating genetically that the Buc-GFP fusion protein is functional. These results show that Buc-GFP exerts all biochemical interactions essential for germline development and highlight the potential of this line to analyze the molecular regulation of germ plasm formation.
Collapse
Affiliation(s)
- Stephan Riemer
- Institut für Entwicklungsbiochemie, GZMB, Universitätsmedizin Göttingen, Georg-August-Universität, Göttingen, Germany
| | - Franck Bontems
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Switzerland
| | - Pritesh Krishnakumar
- Institut für Entwicklungsbiochemie, GZMB, Universitätsmedizin Göttingen, Georg-August-Universität, Göttingen, Germany
| | - Jasmin Gömann
- Institut für Entwicklungsbiochemie, GZMB, Universitätsmedizin Göttingen, Georg-August-Universität, Göttingen, Germany
| | - Roland Dosch
- Institut für Entwicklungsbiochemie, GZMB, Universitätsmedizin Göttingen, Georg-August-Universität, Göttingen, Germany.
| |
Collapse
|
28
|
Desvignes T, Nguyen T, Chesnel F, Bouleau A, Fauvel C, Bobe J. X-Linked Retinitis Pigmentosa 2 Is a Novel Maternal-Effect Gene Required for Left-Right Asymmetry in Zebrafish. Biol Reprod 2015; 93:42. [PMID: 26134862 DOI: 10.1095/biolreprod.115.130575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 06/10/2015] [Indexed: 01/05/2023] Open
Abstract
Retinitis pigmentosa 2 (RP2) gene is responsible for up to 20% of X-linked retinitis pigmentosa, a severe heterogeneous genetic disorder resulting in progressive retinal degeneration in humans. In vertebrates, several bodies of evidence have clearly established the role of Rp2 protein in cilia genesis and/or function. Unexpectedly, some observations in zebrafish have suggested the oocyte-predominant expression of the rp2 gene, a typical feature of maternal-effect genes. In the present study, we investigate the maternal inheritance of rp2 gene products in zebrafish eggs in order to address whether rp2 could be a novel maternal-effect gene required for normal development. Although both rp2 mRNA and corresponding protein are expressed during oogenesis, rp2 mRNA is maternally inherited, in contrast to Rp2 protein. A knockdown of the protein transcribed from both rp2 maternal and zygotic mRNA results in delayed epiboly and severe developmental defects, including eye malformations, that were not observed when only the protein from zygotic origin was knocked down. Moreover, the knockdown of maternal and zygotic Rp2 revealed a high incidence of left-right asymmetry establishment defects compared to only zygotic knockdown. Here we show that rp2 is a novel maternal-effect gene exclusively expressed in oocytes within the zebrafish ovary and demonstrate that maternal rp2 mRNA is essential for successful embryonic development and thus contributes to egg developmental competence. Our observations also reveal that Rp2 protein translated from maternal mRNA is important to allow normal heart loop formation, thus providing evidence of a direct maternal contribution to left-right asymmetry establishment.
Collapse
Affiliation(s)
- Thomas Desvignes
- INRA, UR1037 Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France IFREMER, LALR, Palavas Les Flots, France
| | - Thaovi Nguyen
- INRA, UR1037 Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France
| | | | - Aurélien Bouleau
- INRA, UR1037 Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France IFREMER, LALR, Palavas Les Flots, France
| | | | - Julien Bobe
- INRA, UR1037 Fish Physiology and Genomics, Campus de Beaulieu, Rennes, France
| |
Collapse
|
29
|
Eno C, Pelegri F. Gradual recruitment and selective clearing generate germ plasm aggregates in the zebrafish embryo. BIOARCHITECTURE 2015; 3:125-32. [PMID: 24721731 DOI: 10.4161/bioa.26538] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Determination of primordial germ cells (PGCs) is one of the earliest decisions in animal embryogenesis. In many species, PGCs are determined through maternally-inherited germ plasm ribonucleoparticles (RNPs). In zebrafish, these are transmitted during oogenesis as dispersed RNPs, which after fertilization multimerize and become recruited as large aggregates at furrows for the first and second cell cycles. Here, we show that the number of recruited germ plasm RNPs is halved every cell cycle. We also show that germ plasm RNPs are recruited during the third cell cycle, but only transiently. Our data support a mechanism in which systematic local gathering of germ plasm RNPs during cytokinesis and threshold-dependent clearing contribute to forming germ plasm aggregates with the highest RNP number and germ cell-inducing potential.
Collapse
Affiliation(s)
- Celeste Eno
- Laboratory of Genetics; University of Wisconsin-Madison; Madison, WI USA
| | - Francisco Pelegri
- Laboratory of Genetics; University of Wisconsin-Madison; Madison, WI USA
| |
Collapse
|
30
|
Affiliation(s)
- Julien Bobe
- INRA UR1037, Fish Physiology and Genomics, Rennes F-35000, France
| |
Collapse
|
31
|
Dosch R. Next generation mothers: Maternal control of germline development in zebrafish. Crit Rev Biochem Mol Biol 2014; 50:54-68. [DOI: 10.3109/10409238.2014.985816] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
32
|
Summers AF, Pohlmeier WE, Sargent KM, Cole BD, Vinton RJ, Kurz SG, McFee RM, Cushman RA, Cupp AS, Wood JR. Altered theca and cumulus oocyte complex gene expression, follicular arrest and reduced fertility in cows with dominant follicle follicular fluid androgen excess. PLoS One 2014; 9:e110683. [PMID: 25330369 PMCID: PMC4199720 DOI: 10.1371/journal.pone.0110683] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 09/19/2014] [Indexed: 12/18/2022] Open
Abstract
Aspiration of bovine follicles 12-36 hours after induced corpus luteum lysis serendipitously identified two populations of cows, one with High androstenedione (A4; >40 ng/ml; mean = 102) and another with Low A4 (<20 ng/ml; mean = 9) in follicular fluid. We hypothesized that the steroid excess in follicular fluid of dominant follicles in High A4 cows would result in reduced fertility through altered follicle development and oocyte maternal RNA abundance. To test this hypothesis, estrous cycles of cows were synchronized and ovariectomy was performed 36 hours later. HPLC MS/MS analysis of follicular fluid showed increased dehydroepiandrosterone (6-fold), A4 (158-fold) and testosterone (31-fold) in the dominant follicle of High A4 cows. However, estrone (3-fold) and estradiol (2-fold) concentrations were only slightly elevated, suggesting a possible inefficiency in androgen to estrogen conversion in High A4 cows. Theca cell mRNA expression of LHCGR, GATA6, CYP11A1, and CYP17A1 was greater in High A4 cows. Furthermore, abundance of ZAR1 was decreased 10-fold in cumulus oocyte complexes from High A4 cows, whereas NLRP5 abundance tended to be 19.8-fold greater (P = 0.07). There was a tendency for reduction in stage 4 follicles in ovarian cortex samples from High A4 cows suggesting that progression to antral stages were impaired. High A4 cows tended (P<0.07) to have a 17% reduction in calving rate compared with Low A4 cows suggesting reduced fertility in the High A4 population. These data suggest that the dominant follicle environment of High A4 cows including reduced estrogen conversion and androgen excess contributes to infertility in part through altered follicular and oocyte development.
Collapse
Affiliation(s)
- Adam F. Summers
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - William E. Pohlmeier
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Kevin M. Sargent
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Brizett D. Cole
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Rebecca J. Vinton
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Scott G. Kurz
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Renee M. McFee
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Robert A. Cushman
- USDA-ARS Roman L. Hruska U.S. Meat Animal Research Center, Clay Center, Nebraska, United States of America
| | - Andrea S. Cupp
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
- * E-mail:
| | - Jennifer R. Wood
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| |
Collapse
|
33
|
Kim JD, Jin SW. A tale of two models: mouse and zebrafish as complementary models for lymphatic studies. Mol Cells 2014; 37:503-10. [PMID: 24854860 PMCID: PMC4132301 DOI: 10.14348/molcells.2014.0108] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 05/02/2014] [Indexed: 11/27/2022] Open
Abstract
Lymphatic vessels provide essential roles in maintaining fluid homeostasis and lipid absorption. Dysfunctions of the lymphatic vessels lead to debilitating pathological conditions, collectively known as lymphedema. In addition, lymphatic vessels are a critical moderator for the onset and progression of diverse human diseases including metastatic cancer and obesity. Despite their clinical importance, there is no currently effective pharmacological therapy to regulate functions of lymphatic vessels. Recent efforts to manipulate the Vascular Endothelial Growth Factor-C (VEGFC) pathway, which is arguably the most important signaling pathway regulating lymphatic endothelial cells, to alleviate lymphedema yielded largely mixed results, necessitating identification of new targetable signaling pathways for therapeutic intervention for lymphedema. Zebrafish, a relatively new model system to investigate lymphatic biology, appears to be an ideal model to identify novel therapeutic targets for lymphatic biology. In this review, we will provide an overview of our current understanding of the lymphatic vessels in vertebrates, and discuss zebrafish as a promising in vivo model to study lymphatic vessels.
Collapse
Affiliation(s)
- Jun-Dae Kim
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Suk-Won Jin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
34
|
Leung JYS, Cheung SG, Qiu JW, Ang PO, Chiu JMY, Thiyagarajan V, Shin PKS. Effect of parental hypoxic exposure on embryonic development of the offspring of two serpulid polychaetes: Implication for transgenerational epigenetic effect. MARINE POLLUTION BULLETIN 2013; 74:149-155. [PMID: 23906470 DOI: 10.1016/j.marpolbul.2013.07.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 06/30/2013] [Accepted: 07/04/2013] [Indexed: 06/02/2023]
Abstract
Sperm production and motility, fecundity, and egg size, complexity and viability of serpulid polychaetes Hydroides elegans and Hydroides diramphus after 2-week treatment to hypoxia (2 mg O2 l(-1)) was compared with those under normoxia (6 mg O2 l(-1)). Despite reduced fecundity, the effect of parental hypoxic exposure on gamete quality was not discernible for both species. However, regardless of their subsequent dissolved oxygen environment, eggs spawned by H. elegans after hypoxic exposure were found to have lower fertilization success, slower embryonic development and a significantly higher yield of malformed embryos than those with a parental normoxic treatment. In contrast, neither fertilization success nor rate of embryonic development was affected for H. diramphus. The results implied that hypoxia was a potential stress reducing the recruitment of H. elegans through non-adaptive epigenetic effect, whereas H. diramphus was a more tolerant species to survive hypoxic events.
Collapse
Affiliation(s)
- J Y S Leung
- Department of Biology and Chemistry, City University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
35
|
Herberger AL, Loretz CA. Morpholino oligonucleotide knockdown of the extracellular calcium-sensing receptor impairs early skeletal development in zebrafish. Comp Biochem Physiol A Mol Integr Physiol 2013; 166:470-81. [PMID: 23911792 DOI: 10.1016/j.cbpa.2013.07.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/26/2013] [Accepted: 07/26/2013] [Indexed: 12/15/2022]
Abstract
The complex vertebrate skeleton depends on regulated cell activities to lay down protein matrix and mineral components of bone. As a distinctive vertebrate characteristic, bone is a storage site for physiologically-important calcium ion. The extracellular calcium-sensing receptor (CaSR) is linked to homeostatic regulation of calcium through its expression in endocrine glands that secrete calcium homeostatic hormones, in Ca(2+)- and ion-transporting epithelia, and in skeleton. Since CaSR is restricted in its presence to the chordate-vertebrate evolutionary lineage, we propose there to be important functional ties between CaSRs and vertebrate skeleton in the context of that group's characteristic form of calcium-mineralized skeleton. Since little is known about CaSR in the skeletal biology of non-mammalian vertebrates, reverse transcription-polymerase chain reaction (RT-PCR), in situ hybridization and immunohistochemistry were applied to adult and embryonic zebrafish to reveal CaSR transcript and protein expression in several tissues, including, among these, chondrocytes and developing bone and notochord as components in skeletal development. Morpholino oligonucleotide (MO) knockdown technique was used to probe CaSR role(s) in the zebrafish model system. By RT-PCR assessment, injection of a splice-inhibiting CaSR MO reduced normally-spliced Casr gene transcript expression measured at 2days postfertilization (dpf). Corresponding to the knockdown of normally-spliced mRNA by the CaSR MO, we observed a morphant phenotype characterized by stunted growth and disorganization of the notochord and axial skeleton by 1dpf. We conclude that, like its critically important role in normal bone development in mammals, CaSR is essential in skeletogenesis in fishes.
Collapse
Affiliation(s)
- Amanda L Herberger
- Department of Biological Sciences, University at Buffalo, Buffalo, NY 14260-1300, USA.
| | | |
Collapse
|
36
|
Nair S, Marlow F, Abrams E, Kapp L, Mullins MC, Pelegri F. The chromosomal passenger protein birc5b organizes microfilaments and germ plasm in the zebrafish embryo. PLoS Genet 2013; 9:e1003448. [PMID: 23637620 PMCID: PMC3630083 DOI: 10.1371/journal.pgen.1003448] [Citation(s) in RCA: 36] [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: 07/13/2012] [Accepted: 02/28/2013] [Indexed: 12/21/2022] Open
Abstract
Microtubule-microfilament interactions are important for cytokinesis and subcellular localization of proteins and mRNAs. In the early zebrafish embryo, astral microtubule-microfilament interactions also facilitate a stereotypic segregation pattern of germ plasm ribonucleoparticles (GP RNPs), which is critical for their eventual selective inheritance by germ cells. The precise mechanisms and molecular mediators for both cytoskeletal interactions and GP RNPs segregation are the focus of intense research. Here, we report the molecular identification of a zebrafish maternal-effect mutation motley as Birc5b, a homolog of the mammalian Chromosomal Passenger Complex (CPC) component Survivin. The meiosis and mitosis defects in motley/birc5b mutant embryos are consistent with failed CPC function, and additional defects in astral microtubule remodeling contribute to failures in the initiation of cytokinesis furrow ingression. Unexpectedly, the motley/birc5b mutation also disrupts cortical microfilaments and GP RNP aggregation during early cell divisions. Birc5b localizes to the tips of astral microtubules along with polymerizing cortical F-actin and the GP RNPs. Mutant Birc5b co-localizes with cortical F-actin and GP RNPs, but fails to associate with astral microtubule tips, leading to disorganized microfilaments and GP RNP aggregation defects. Thus, maternal Birc5b localizes to astral microtubule tips and associates with cortical F-actin and GP RNPs, potentially linking the two cytoskeletons to mediate microtubule-microfilament reorganization and GP RNP aggregation during early embryonic cell cycles in zebrafish. In addition to the known mitotic function of CPC components, our analyses reveal a non-canonical role for an evolutionarily conserved CPC protein in microfilament reorganization and germ plasm aggregation. We address mechanisms by which germ cell precursors, a cell type that generates sperm and eggs for future generations, are specified in the zebrafish. Germ cell-specific genes are highly conserved across species, and in many animals germ cells are specified by the inheritance of germ plasm, a specialized cytoplasm containing specific proteins and RNAs corresponding to such conserved genes. Germ plasm is inherited as ribonucleoparticles, which are often present in the egg as singletons and which aggregate to generate larger masses that, when inherited by germ cell precursors, will initiate a germ cell-specific gene expression program. Here, we present the functional and molecular analysis of the zebrafish maternal gene, motley, which we show encodes a homologue of the Chromosomal Passenger Complex protein Survivin, or Birc5b. We found that, in addition to the expected role of this protein in cell division, characteristic of factors in this complex, Birc5b mediates germ plasm aggregation in the early zebrafish embryo through the coordination of dynamic changes in the cytoskeleton. Our studies provide a mechanistic basis to explain how germ cell determinants are transmitted from one generation to the next and reveal a non-conventional role for a Chromosomal Passenger Complex factor in this process.
Collapse
Affiliation(s)
- Sreelaja Nair
- Laboratory of Genetics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Florence Marlow
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elliott Abrams
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Lee Kapp
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mary C. Mullins
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Francisco Pelegri
- Laboratory of Genetics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
37
|
|
38
|
Nair S, Lindeman RE, Pelegri F. In vitro oocyte culture-based manipulation of zebrafish maternal genes. Dev Dyn 2012; 242:44-52. [PMID: 23074011 DOI: 10.1002/dvdy.23894] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2012] [Indexed: 12/12/2022] Open
Abstract
In animals, females deposit gene products into developing oocytes, which drive early cellular events in embryos immediately after fertilization. As maternal gene products are present before fertilization, the functional manipulation of maternal genes is often challenging to implement, requiring gene expression or targeting during oogenesis. Maternal expression can be achieved through transgenesis, but transgenic approaches are time consuming and subject to undesired epigenetic effects. Here, we have implemented in vitro culturing of experimentally manipulated immature oocytes to study maternal gene contribution to early embryonic development in the zebrafish. We demonstrate phenotypic rescue of a maternal-effect mutation by expressing wild-type product in cultured oocytes. We also generate loss-of-function phenotypes in embryos through either the expression of a dominant-negative transcript or injection of translation-blocking morpholino oligonucleotides. Finally, we demonstrate subcellular localization during the early cell divisions immediately after fertilization of an exogenously provided maternal product fused to a fluorescent protein. These manipulations extend the potential to carry out genetic and imaging studies of zebrafish maternal genes during the egg-to-embryo transition.
Collapse
Affiliation(s)
- Sreelaja Nair
- Laboratory of Genetics, University of Wisconsin - Madison, Madison, WI, USA
| | | | | |
Collapse
|
39
|
Law SHW, Sargent TD. Maternal pak4 expression is required for primitive myelopoiesis in zebrafish. Mech Dev 2012; 130:181-94. [PMID: 23032194 DOI: 10.1016/j.mod.2012.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 09/21/2012] [Indexed: 01/08/2023]
Abstract
Transcripts of pak4, the zebrafish ortholog of p21-activated kinase 4 (PAK4), are most abundant in the egg and fall to low levels by the end of gastrulation, after which expression is essentially ubiquitous. Translation of maternal mRNA into pak4 protein is first detectable at high stage (3.3hpf). Splice-blocking morpholino oligonucleotides (MOs) were used to prevent zygotic pak4 expression. This had no discernable effect on development through larval stages. In contrast, a translation-blocking MO, alone or in combination with the splice MOs, resulted in a complex lethal phenotype. In addition to disrupted somite development and other morphogenetic abnormalities, the knockdown of maternal pak4 expression led to alterations in regulatory gene expression in the primitive hematopoietic domains, leading to deficiencies in granulocyte and leukocyte lineages. At least some of the effects of pak4 knockdown on gene expression could be mimicked by treatment with actin depolymerization agents, suggesting a mechanistic link between regulation of microfilament dynamics by pak4 and regulation of gene expression in primitive myeloid cell differentiation.
Collapse
Affiliation(s)
- Sheran H W Law
- Section on Vertebrate Development, Program on Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Development, National Institutes of Health, Bethesda, MD, USA
| | | |
Collapse
|
40
|
Centrosomes in the zebrafish (Danio rerio): a review including the related basal body. Cilia 2012; 1:9. [PMID: 23351173 PMCID: PMC3555702 DOI: 10.1186/2046-2530-1-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 06/07/2012] [Indexed: 12/15/2022] Open
Abstract
Ever since Edouard Van Beneden and Theodor Boveri first formally described the centrosome in the late 1800s, it has captivated cell biologists. The name clearly indicated its central importance to cell functioning, even to these early investigators. We now know of its role as a major microtubule-organizing center (MTOC) and of its dynamic roles in cell division, vesicle trafficking and for its relative, the basal body, ciliogenesis. While centrosomes are found in most animal cells, notably it is absent in most oocytes and higher plant cells. Nevertheless, it appears that critical components of the centrosome act as MTOCs in these cells as well. The zebrafish has emerged as an exciting and promising new model organism, primarily due to the pioneering efforts of George Streisinger to use zebrafish in genetic studies and due to Christiane Nusslein-Volhard, Wolfgang Driever and their teams of collaborators, who applied forward genetics to elicit a large number of mutant lines. The transparency and rapid external development of the embryo allow for experiments not easily done in other vertebrates. The ease of producing transgenic lines, often with the use of fluorescent reporters, and gene knockdowns with antisense morpholinos further contributes to the appeal of the model as an experimental system. The added advantage of high-throughput screening of small-molecule libraries, as well as the ease of mass rearing together with low cost, makes the zebrafish a true frontrunner as a model vertebrate organism. The zebrafish has a body plan shared by all vertebrates, including humans. This conservation of body plan provides added significance to the existing lines of zebrafish as human disease models and adds an impetus to the ongoing efforts to develop new models. In this review, the current state of knowledge about the centrosome in the zebrafish model is explored. Also, studies on the related basal body in zebrafish and their relationship to ciliogenesis are reviewed.
Collapse
|
41
|
Ho DH, Burggren WW. Parental hypoxic exposure confers offspring hypoxia resistance in zebrafish (Danio rerio). J Exp Biol 2012; 215:4208-16. [DOI: 10.1242/jeb.074781] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Summary
Maternal influences are a potentially important component of transgenerational transfer of phenotype in vertebrates. This study on zebrafish (Danio rerio) examined how chronic hypoxic exposure on adults affected the phenotype of their offspring. Separate adult populations were exposed to hypoxia (13.1 kPa O2) or normoxia (21.1 kPa O2) for periods ranging from 1 to 12 weeks. Adults were then returned to normoxia and bred within experimental groups. Adult fecundity and egg characteristics (volume of egg, yolk and perivitelline fluid) were assessed. Subsequently, larval body length, time to loss of equilibrium in severe hypoxia (~4 kPa O2), and critical thermal minima (CTMin) and maxima (CTMax) were measured at 6, 9, 12, 15, 18, 21 and 60 days post fertilization (dpf). Adult fecundity was depressed by hypoxic exposure. Egg component volumes were also depressed in adults exposed to 1-2 weeks of hypoxia, but returned to control levels following longer hypoxic exposure. Adult hypoxic exposures of >1 week resulted in longer body lengths in their larval offspring. Time to loss of equilibrium in severe hypoxia (i.e. hypoxic resistance) in control larvae decreased from 6 to 12 dpf, remaining constant thereafter. Notably, hypoxic resistance from 6-18 dpf was ~15% lower in larvae whose parents were exposed to just 1 week of chronic hypoxia, but resistance was significantly increased by ~24-30% in 6-18 dpf in larvae from adults exposed to 2, 3 or 4 weeks of hypoxia. CTMin (~39.5°C) and CTMax (~10-12 °C) were unchanged by parental hypoxic exposure. This study demonstrates that parental hypoxic exposure in adult zebrafish has profound epigenetic effects on the morphological and physiological phenotype of their offspring.
Collapse
Affiliation(s)
- Dao H. Ho
- Georgia Health Sciences University, Augusta, USA; University of North Texas, Denton, USA
| | | |
Collapse
|
42
|
Salinas-Saavedra M, Vargas AO. Cortical cytasters: a highly conserved developmental trait of Bilateria with similarities to Ctenophora. EvoDevo 2011; 2:23. [PMID: 22133482 PMCID: PMC3248832 DOI: 10.1186/2041-9139-2-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 12/01/2011] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Cytasters (cytoplasmic asters) are centriole-based nucleation centers of microtubule polymerization that are observable in large numbers in the cortical cytoplasm of the egg and zygote of bilaterian organisms. In both protostome and deuterostome taxa, cytasters have been described to develop during oogenesis from vesicles of nuclear membrane that move to the cortical cytoplasm. They become associated with several cytoplasmic components, and participate in the reorganization of cortical cytoplasm after fertilization, patterning the antero-posterior and dorso-ventral body axes. PRESENTATION OF THE HYPOTHESIS The specific resemblances in the development of cytasters in both protostome and deuterostome taxa suggest that an independent evolutionary origin is unlikely. An assessment of published data confirms that cytasters are present in several protostome and deuterostome phyla, but are absent in the non-bilaterian phyla Cnidaria and Ctenophora. We hypothesize that cytasters evolved in the lineage leading to Bilateria and were already present in the most recent common ancestor shared by protostomes and deuterostomes. Thus, cytasters would be an ancient and highly conserved trait that is homologous across the different bilaterian phyla. The alternative possibility is homoplasy, that is cytasters have evolved independently in different lineages of Bilateria. TESTING THE HYPOTHESIS So far, available published information shows that appropriate observations have been made in eight different bilaterian phyla. All of them present cytasters. This is consistent with the hypothesis of homology and conservation. However, there are several important groups for which there are no currently available data. The hypothesis of homology predicts that cytasters should be present in these groups. Increasing the taxonomic sample using modern techniques uniformly will test for evolutionary patterns supporting homology, homoplasy, or secondary loss of cytasters. IMPLICATIONS OF THE HYPOTHESIS If cytasters are homologous and highly conserved across bilateria, their potential developmental and evolutionary relevance has been underestimated. The deep evolutionary origin of cytasters also becomes a legitimate topic of research. In Ctenophora, polyspermic fertilization occurs, with numerous sperm entering the egg. The centrosomes of sperm pronuclei associate with cytoplasmic components of the egg and reorganize the cortical cytoplasm, defining the oral-aboral axis. These resemblances lead us to suggest the possibility of a polyspermic ancestor in the lineage leading to Bilateria.
Collapse
Affiliation(s)
- Miguel Salinas-Saavedra
- Laboratory of Ontogeny and Phylogeny, Department of Biology, Faculty of Science, University of Chile. Las Palmeras, Ñuñoa, Casilla 653, Santiago, Chile
| | - Alexander O Vargas
- Laboratory of Ontogeny and Phylogeny, Department of Biology, Faculty of Science, University of Chile. Las Palmeras, Ñuñoa, Casilla 653, Santiago, Chile
| |
Collapse
|
43
|
Kirioukhova O, Johnston AJ, Kleen D, Kägi C, Baskar R, Moore JM, Bäumlein H, Gross-Hardt R, Grossniklaus U. Female gametophytic cell specification and seed development require the function of the putative Arabidopsis INCENP ortholog WYRD. Development 2011; 138:3409-20. [PMID: 21752930 DOI: 10.1242/dev.060384] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In plants, gametes, along with accessory cells, are formed by the haploid gametophytes through a series of mitotic divisions, cell specification and differentiation events. How the cells in the female gametophyte of flowering plants differentiate into gametes (the egg and central cell) and accessory cells remains largely unknown. In a screen for mutations that affect egg cell differentiation in Arabidopsis, we identified the wyrd (wyr) mutant, which produces additional egg cells at the expense of the accessory synergids. WYR not only restricts gametic fate in the egg apparatus, but is also necessary for central cell differentiation. In addition, wyr mutants impair mitotic divisions in the male gametophyte and endosperm, and have a parental effect on embryo cytokinesis, consistent with a function of WYR in cell cycle regulation. WYR is upregulated in gametic cells and encodes a putative plant ortholog of the inner centromere protein (INCENP), which is implicated in the control of chromosome segregation and cytokinesis in yeast and animals. Our data reveal a novel developmental function of the conserved cell cycle-associated INCENP protein in plant reproduction, in particular in the regulation of egg and central cell fate and differentiation.
Collapse
Affiliation(s)
- Olga Kirioukhova
- Institute of Plant Biology and Zürich-Basel Plant Science Center, University of Zürich, Zürich, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Aanes H, Winata CL, Lin CH, Chen JP, Srinivasan KG, Lee SGP, Lim AYM, Hajan HS, Collas P, Bourque G, Gong Z, Korzh V, Aleström P, Mathavan S. Zebrafish mRNA sequencing deciphers novelties in transcriptome dynamics during maternal to zygotic transition. Genome Res 2011; 21:1328-38. [PMID: 21555364 DOI: 10.1101/gr.116012.110] [Citation(s) in RCA: 226] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Maternally deposited mRNAs direct early development before the initiation of zygotic transcription during mid-blastula transition (MBT). To study mechanisms regulating this developmental event in zebrafish, we applied mRNA deep sequencing technology and generated comprehensive information and valuable resources on transcriptome dynamics during early embryonic (egg to early gastrulation) stages. Genome-wide transcriptome analysis documented at least 8000 maternal genes and identified the earliest cohort of zygotic transcripts. We determined expression levels of maternal and zygotic transcripts with the highest resolution possible using mRNA-seq and clustered them based on their expression pattern. We unravel delayed polyadenylation in a large cohort of maternal transcripts prior to the MBT for the first time in zebrafish. Blocking polyadenylation of these transcripts confirms their role in regulating development from the MBT onward. Our study also identified a large number of novel transcribed regions in annotated and unannotated regions of the genome, which will facilitate reannotation of the zebrafish genome. We also identified splice variants with an estimated frequency of 50%-60%. Taken together, our data constitute a useful genomic information and valuable transcriptome resource for gene discovery and for understanding the mechanisms of early embryogenesis in zebrafish.
Collapse
Affiliation(s)
- Håvard Aanes
- BasAM, Norwegian School of Veterinary Science, 0033 Oslo, Norway
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
McGinnis LK, Hong X, Christenson LK, Kinsey WH. Fer tyrosine kinase is required for germinal vesicle breakdown and meiosis-I in mouse oocytes. Mol Reprod Dev 2011; 78:33-47. [PMID: 21268181 DOI: 10.1002/mrd.21264] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The control of microtubule and actin-mediated events that direct the physical arrangement and separation of chromosomes during meiosis is critical since failure to maintain chromosome organization can lead to germ cell aneuploidy. Our previous studies demonstrated a role for FYN tyrosine kinase in chromosome and spindle organization and in cortical polarity of the mature mammalian oocyte. In addition to Fyn, mammalian oocytes express the protein tyrosine kinase Fer at high levels relative to other tissues. The objective of the present study was to determine the function of this kinase in the oocyte. Feline encephalitis virus (FES)-related kinase (FER) protein was uniformly distributed in the ooplasm of small oocytes, but became concentrated in the germinal vesicle (GV) during oocyte growth. After germinal vesicle breakdown (GVBD), FER associated with the metaphase-I (MI) and metaphase-II (MII) spindles. Suppression of Fer expression by siRNA knockdown in GV stage oocytes did not prevent activation of cyclin dependent kinase 1 activity or chromosome condensation during in vitro maturation, but did arrest oocytes prior to GVBD or during MI. The resultant phenotype displayed condensed chromosomes trapped in the GV, or condensed chromosomes poorly arranged in a metaphase plate but with an underdeveloped spindle microtubule structure or chromosomes compacted into a tight sphere. The results demonstrate that FER kinase plays a critical role in oocyte meiotic spindle microtubule dynamics and may have an additional function in GVBD.
Collapse
Affiliation(s)
- Lynda K McGinnis
- Department of Anatomy and Cell Biology, University of Kansas Medical School, Kansas City, Kansas 66160, USA.
| | | | | | | |
Collapse
|
46
|
Rodríguez-Marí A, Wilson C, Titus TA, Cañestro C, BreMiller RA, Yan YL, Nanda I, Johnston A, Kanki JP, Gray EM, He X, Spitsbergen J, Schindler D, Postlethwait JH. Roles of brca2 (fancd1) in oocyte nuclear architecture, gametogenesis, gonad tumors, and genome stability in zebrafish. PLoS Genet 2011; 7:e1001357. [PMID: 21483806 PMCID: PMC3069109 DOI: 10.1371/journal.pgen.1001357] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 02/28/2011] [Indexed: 01/07/2023] Open
Abstract
Mild mutations in BRCA2 (FANCD1) cause Fanconi anemia (FA) when homozygous, while severe mutations cause common cancers including breast, ovarian, and prostate cancers when heterozygous. Here we report a zebrafish brca2 insertional mutant that shares phenotypes with human patients and identifies a novel brca2 function in oogenesis. Experiments showed that mutant embryos and mutant cells in culture experienced genome instability, as do cells in FA patients. In wild-type zebrafish, meiotic cells expressed brca2; and, unexpectedly, transcripts in oocytes localized asymmetrically to the animal pole. In juvenile brca2 mutants, oocytes failed to progress through meiosis, leading to female-to-male sex reversal. Adult mutants became sterile males due to the meiotic arrest of spermatocytes, which then died by apoptosis, followed by neoplastic proliferation of gonad somatic cells that was similar to neoplasia observed in ageing dead end (dnd)-knockdown males, which lack germ cells. The construction of animals doubly mutant for brca2 and the apoptotic gene tp53 (p53) rescued brca2-dependent sex reversal. Double mutants developed oocytes and became sterile females that produced only aberrant embryos and showed elevated risk for invasive ovarian tumors. Oocytes in double-mutant females showed normal localization of brca2 and pou5f1 transcripts to the animal pole and vasa transcripts to the vegetal pole, but had a polarized rather than symmetrical nucleus with the distribution of nucleoli and chromosomes to opposite nuclear poles; this result revealed a novel role for Brca2 in establishing or maintaining oocyte nuclear architecture. Mutating tp53 did not rescue the infertility phenotype in brca2 mutant males, suggesting that brca2 plays an essential role in zebrafish spermatogenesis. Overall, this work verified zebrafish as a model for the role of Brca2 in human disease and uncovered a novel function of Brca2 in vertebrate oocyte nuclear architecture. Women with one strong BRCA2(FANCD1) mutation have high risks of breast and ovarian cancer. People with two mild BRCA2(FANCD1) mutations develop Fanconi Anemia, which reduces DNA repair leading to genome instability, small gonads, infertility, and cancer. Humans and mice lacking BRCA2 activity die before birth. We discovered that zebrafish brca2 mutants show chromosome instability and small gonads, and they develop only as sterile adult males. Female-to-male sex reversal is due to oocyte death during sex determination. Normal animals expressed brca2 in developing eggs and sperm that are repairing DNA breaks associated with genetic reshuffling. Normal developing eggs localized brca2 RNA near the nucleus, suggesting a role in protecting rapidly dividing early embryonic cells. Sperm-forming cells died in adult mutant males. Inhibition of cell death rescued sex reversal, but not fertility. Rescued females developed invasive ovarian tumors and formed eggs with abnormal nuclear architecture. The novel role of Brca2 in organizing the vertebrate egg nucleus may provide new insights into the origin of ovarian cancer. These results validate zebrafish as a model for human BRCA2-related diseases and provide a tool for the identification of substances that can rescue zebrafish brca2 mutants and thus become candidates for therapeutic molecules for human disease.
Collapse
Affiliation(s)
- Adriana Rodríguez-Marí
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Catherine Wilson
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Tom A. Titus
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Cristian Cañestro
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Ruth A. BreMiller
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Yi-Lin Yan
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Indrajit Nanda
- Institute of Human Genetics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Adam Johnston
- Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - John P. Kanki
- Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Erin M. Gray
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Xinjun He
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
| | - Jan Spitsbergen
- Marine and Freshwater Biomedical Sciences Center, Oregon State University, Corvallis, Oregon, United States of America
| | - Detlev Schindler
- Institute of Human Genetics, Biocenter, University of Würzburg, Würzburg, Germany
| | - John H. Postlethwait
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, United States of America
- * E-mail:
| |
Collapse
|
47
|
Wühr M, Obholzer N, Megason S, Detrich H, Mitchison T. Live imaging of the cytoskeleton in early cleavage-stage zebrafish embryos. Methods Cell Biol 2011; 101:1-18. [PMID: 21550437 PMCID: PMC6551615 DOI: 10.1016/b978-0-12-387036-0.00001-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The large and transparent cells of cleavage-stage zebrafish embryos provide unique opportunities to study cell division and cytoskeletal dynamics in very large animal cells. Here, we summarize recent progress, from our laboratories and others, on live imaging of the microtubule and actin cytoskeletons during zebrafish embryonic cleavage. First, we present simple protocols for extending the breeding competence of zebrafish mating ensembles throughout the day, which ensures a steady supply of embryos in early cleavage, and for mounting these embryos for imaging. Second, we describe a transgenic zebrafish line [Tg(bactin2:HsENSCONSIN17-282-3xEGFP)hm1] that expresses the green fluorescent protein (GFP)-labeled microtubule-binding part of ensconsin (EMTB-3GFP). We demonstrate that the microtubule-based structures of the early cell cycles can be imaged live, with single microtubule resolution and with high contrast, in this line. Microtubules are much more easily visualized using this tagged binding protein rather than directly labeled tubulin (injected Alexa-647-labeled tubulin), presumably due to lower background from probe molecules not attached to microtubules. Third, we illustrate live imaging of the actin cytoskeleton by injection of the actin-binding fragment of utrophin fused to GFP. Fourth, we compare epifluorescence-, spinning-disc-, laser-scanning-, and two-photon-microscopic modalities for live imaging of the microtubule cytoskeleton in early embryos of our EMTB-3GFP-expressing transgenic line. Finally, we discuss future applications and extensions of our methods.
Collapse
Affiliation(s)
- M. Wühr
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - N.D. Obholzer
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - S.G. Megason
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - H.W. Detrich
- Department of Biology, Northeastern University, Boston, Massachusetts 02115, USA
| | - T.J. Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
48
|
Pelegri F, Mullins MC. Genetic Screens for Mutations Affecting Adult Traits and Parental-effect Genes. Methods Cell Biol 2011; 104:83-120. [DOI: 10.1016/b978-0-12-374814-0.00005-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
49
|
Böhne A, Darras A, D'Cotta H, Baroiller JF, Galiana-Arnoux D, Volff JN. The vertebrate makorin ubiquitin ligase gene family has been shaped by large-scale duplication and retroposition from an ancestral gonad-specific, maternal-effect gene. BMC Genomics 2010; 11:721. [PMID: 21172006 PMCID: PMC3022923 DOI: 10.1186/1471-2164-11-721] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 12/20/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Members of the makorin (mkrn) gene family encode RING/C3H zinc finger proteins with U3 ubiquitin ligase activity. Although these proteins have been described in a variety of eukaryotes such as plants, fungi, invertebrates and vertebrates including human, almost nothing is known about their structural and functional evolution. RESULTS Via partial sequencing of a testis cDNA library from the poeciliid fish Xiphophorus maculatus, we have identified a new member of the makorin gene family, that we called mkrn4. In addition to the already described mkrn1 and mkrn2, mkrn4 is the third example of a makorin gene present in both tetrapods and ray-finned fish. However, this gene was not detected in mouse and rat, suggesting its loss in the lineage leading to rodent murids. Mkrn2 and mkrn4 are located in large ancient duplicated regions in tetrapod and fish genomes, suggesting the possible involvement of ancestral vertebrate-specific genome duplication in the formation of these genes. Intriguingly, many mkrn1 and mkrn2 intronless retrocopies have been detected in mammals but not in other vertebrates, most of them corresponding to pseudogenes. The nature and number of zinc fingers were found to be conserved in Mkrn1 and Mkrn2 but much more variable in Mkrn4, with lineage-specific differences. RT-qPCR analysis demonstrated a highly gonad-biased expression pattern for makorin genes in medaka and zebrafish (ray-finned fishes) and amphibians, but a strong relaxation of this specificity in birds and mammals. All three mkrn genes were maternally expressed before zygotic genome activation in both medaka and zebrafish early embryos. CONCLUSION Our analysis demonstrates that the makorin gene family has evolved through large-scale duplication and subsequent lineage-specific retroposition-mediated duplications in vertebrates. From the three major vertebrate mkrn genes, mkrn4 shows the highest evolutionary dynamics, with lineage-specific loss of zinc fingers and even complete gene elimination from certain groups of vertebrates. Comparative expression analysis strongly suggests that the ancestral E3 ubiquitin ligase function of the single copy mkrn gene before duplication in vertebrates was gonad-specific, with maternal expression in early embryos.
Collapse
Affiliation(s)
- Astrid Böhne
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, Lyon, France.
| | | | | | | | | | | |
Collapse
|
50
|
Pereira M, Costa F, Oliveira A, Serapião R, Machado M, Viana JM, Camargo L. Quantificação de transcritos maternos em oócitos bovinos submetidos a diferentes condições de maturação. ARQ BRAS MED VET ZOO 2010. [DOI: 10.1590/s0102-09352010000600015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Comparou-se a quantidade relativa de transcritos de origem materna entre oócitos bovinos maturados in vivo e maturados em diferentes condições in vitro. Avaliou-se também o efeito dos sistemas de maturação in vitro sobre a viabilidade das células do cumulus. Para a maturação in vivo, os oócitos foram coletados 19-20h após aplicação de gonadorelina em doadoras superestimuladas com FSH e sincronizadas com implante de progesterona. Para a maturação in vitro, oócitos imaturos, obtidos de ovários coletados em matadouro, foram maturados sob diferentes tensões de oxigênio e suplementação proteica. Avaliou-se a abundância dos transcritos de Zar1, MATER e GDF9 por PCR em tempo real. A viabilidade das células do cumulus de oócitos maturados in vitro foi analisada pela coloração de Azul de Tripan. Observou-se sub-regulação (P<0,05) dos transcritos em oócitos submetidos às diferentes condições de maturação in vitro em relação aos maturados in vivo. Não houve diferença (P>0,05) na viabilidade das células do cumulus. Conclui-se que o sistema de maturação influencia a quantidade de transcritos de origem materna armazenados no citoplasma de oócitos bovinos
Collapse
Affiliation(s)
- M.M Pereira
- Embrapa Gado de Leite; Universidade Federal de Juiz de Fora
| | | | | | | | - M.A Machado
- Embrapa Gado de Leite; Universidade Federal de Juiz de Fora
| | | | | |
Collapse
|