1
|
Li YR, Ling LB, Chao A, Fugmann SD, Yang SY. Transient chromatin decompaction at the start of D. melanogaster male embryonic germline development. Life Sci Alliance 2024; 7:e202302401. [PMID: 38991729 PMCID: PMC11239976 DOI: 10.26508/lsa.202302401] [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: 09/27/2023] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/13/2024] Open
Abstract
Embryonic germ cells develop rapidly to establish the foundation for future developmental trajectories, and in this process, they make critical lineage choices including the configuration of their unique identity and a decision on sex. Here, we use single-cell genomics patterns for the entire embryonic germline in Drosophila melanogaster along with the somatic gonadal precursors after embryonic gonad coalescence to investigate molecular mechanisms involved in the setting up and regulation of the germline program. Profiling of the early germline chromatin landscape revealed sex- and stage-specific features. In the male germline immediately after zygotic activation, the chromatin structure underwent a brief remodeling phase during which nucleosome density was lower and deconcentrated from promoter regions. These findings echoed enrichment analysis results of our genomics data in which top candidates were factors with the ability to mediate large-scale chromatin reorganization. Together, they point to the importance of chromatin regulation in the early germline and raise the possibility of a conserved epigenetic reprogramming-like process required for proper initiation of germline development.
Collapse
Affiliation(s)
- Yi-Ru Li
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li Bin Ling
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Angel Chao
- Department of Obstetrics and Gynecology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Sebastian D Fugmann
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu Yuan Yang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Obstetrics and Gynecology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
2
|
Mahadevaraju S, Pal S, Bhaskar P, McDonald BD, Benner L, Denti L, Cozzi D, Bonizzoni P, Przytycka TM, Oliver B. Diverse somatic Transformer and sex chromosome karyotype pathways regulate gene expression in Drosophila gonad development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607556. [PMID: 39372789 PMCID: PMC11451611 DOI: 10.1101/2024.08.12.607556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The somatic sex determination gene transformer (tra) is required for the highly sexually dimorphic development of most somatic cells, including those of the gonads. In addition, somatic tra is required for the germline development even though it is not required for sex determination within germ cells. Germ cell autonomous gene expression is also necessary for their sex determination. To understand the interplay between these signals, we compared the phenotype and gene expression of larval wild-type gonads and the sex-transformed tra gonads. XX larval ovaries transformed into testes were dramatically smaller than wild-type, with significant reductions in germ cell number, likely due to altered geometry of the stem cell niche. Additionally, there was a defect in progression into spermatocyte stages. XY larval testes transformed into ovaries had excessive germ cells, possibly due to the earlier onset of cell division. We suggest that germ cells are neither fully female nor male following somatic sex transformation, with certain pathways characteristic of each sex expressed in tra mutants. We found multiple patterns of somatic and germline gene expression control exclusively due to tra, exclusively due to sex chromosome karyotype, but usually due to a combination of these factors showing tra and sex chromosome karyotype pathways regulate gene expression during Drosophila gonad development.
Collapse
Affiliation(s)
- Sharvani Mahadevaraju
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Biology. St. Mary’s College of Maryland, St. Mary’s City, Maryland, USA
| | - Soumitra Pal
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
- Neurobiology Neurodegeneration and Repair Lab, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Pradeep Bhaskar
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Brennan D. McDonald
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Biology, Stanford University, Stanford, California, USA
| | - Leif Benner
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Luca Denti
- Department of Informatics, Systems, and Communication, University of Milano - Bicocca, Milan, Italy
| | - Davide Cozzi
- Department of Informatics, Systems, and Communication, University of Milano - Bicocca, Milan, Italy
| | - Paola Bonizzoni
- Department of Informatics, Systems, and Communication, University of Milano - Bicocca, Milan, Italy
| | - Teresa M. Przytycka
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian Oliver
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Zhang R, Shi P, Xu S, Ming Z, Liu Z, He Y, Dai J, Matunis E, Xu J, Ma Q. Soma-germline communication drives sex maintenance in the Drosophila testis. Natl Sci Rev 2024; 11:nwae215. [PMID: 39183747 PMCID: PMC11342250 DOI: 10.1093/nsr/nwae215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 08/27/2024] Open
Abstract
In adult gonads, disruption of somatic sexual identity leads to defective gametogenesis and infertility. However, the underlying mechanisms by which somatic signals regulate germline cells to achieve proper gametogenesis remain unclear. In our previous study, we introduced the chinmoSex Transformation (chinmoST ) mutant Drosophila testis phenotype as a valuable model for investigating the mechanisms underlying sex maintenance. In chinmoST testes, depletion of the Janus Kinase-Signal Transducer and Activator of Transcription downstream effector Chinmo from somatic cyst stem cells (CySCs) feminizes somatic cyst cells and arrests germline differentiation. Here, we use single-cell RNA sequencing to uncover chinmoST -specific cell populations and their transcriptomic changes during sex transformation. Comparative analysis of intercellular communication networks between wild-type and chinmoST testes revealed disruptions in several soma-germline signaling pathways in chinmoST testes. Notably, the insulin signaling pathway exhibited significant enhancement in germline stem cells (GSCs). Chinmo cleavage under targets and tagmentation (CUT&Tag) assay revealed that Chinmo directly regulates two male sex determination factors, doublesex (dsx) and fruitless (fru), as well as Ecdysone-inducible gene L2 (ImpL2), a negative regulator of the insulin signaling pathway. Further genetic manipulations confirmed that the impaired gametogenesis observed in chinmoST testes was partly contributed by dysregulation of the insulin signaling pathway. In summary, our study demonstrates that somatic sex maintenance promotes normal spermatogenesis through Chinmo-mediated conserved sex determination and the insulin signaling pathway. Our work offers new insights into the complex mechanisms of somatic stem cell sex maintenance and soma-germline communication at the single-cell level. Additionally, our discoveries highlight the potential significance of stem cell sex instability as a novel mechanism contributing to testicular tumorigenesis.
Collapse
Affiliation(s)
- Rui Zhang
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Peiyu Shi
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Shuyang Xu
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhe Ming
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zicong Liu
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuanyuan He
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Junbiao Dai
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Erika Matunis
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Jin Xu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Qing Ma
- Shenzhen Key Laboratory of Synthetic Genomics, Guangdong Provincial Key Laboratory of Synthetic Genomics, Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
4
|
Ota R, Miura H, Masukawa M, Hayashi M, Kobayashi S. Identification of novel candidate genes leading to sex differentiation in primordial germ cells of Drosophila. Gene Expr Patterns 2023; 48:119321. [PMID: 37142099 DOI: 10.1016/j.gep.2023.119321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 03/28/2023] [Accepted: 04/26/2023] [Indexed: 05/06/2023]
Abstract
Germline sex determination and differentiation are pivotal processes in reproduction. In Drosophila, sex determination of the germline occurs in primordial germ cells (PGCs), and the sex differentiation of these cells is initiated during embryogenesis. However, the molecular mechanism initiating sex differentiation remains elusive. To address this issue, we identified sex-biased genes using RNA-sequencing data of male and female PGCs. Our research revealed 497 genes that were differentially expressed more than twofold between sexes and expressed at high or moderate levels in either male or female PGCs. Among these genes, we used microarray data of PGCs and whole embryos to select 33 genes, which are predominantly expressed in PGCs compared to the soma, as candidate genes contributing to sex differentiation. Of 497 genes, 13 genes that were differentially expressed more than fourfold between sexes were also selected as candidates. Among the 46 (33 + 13) candidates, we confirmed the sex-biased expression of 15 genes by in situ hybridization and quantitative reverse transcription-polymerase chain reaction (qPCR) analysis. Six and nine genes were predominantly expressed in male and female PGCs, respectively. These results represent a first step toward elucidating the mechanisms that initiate sex differentiation in the germline.
Collapse
Affiliation(s)
- Ryoma Ota
- Department of Biosciences, Faculty of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, 320-8551, Japan; Division of Integrated Science and Engineering, Graduate School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi, 320-8551, Japan.
| | - Hiroki Miura
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Masaki Masukawa
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Makoto Hayashi
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Satoru Kobayashi
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan; Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan; Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
5
|
Masculinizer and Doublesex as Key Factors Regulate Sexual Dimorphism in Ostrinia furnacalis. Cells 2022; 11:cells11142161. [PMID: 35883604 PMCID: PMC9320909 DOI: 10.3390/cells11142161] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/06/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary In animals, sexually dimorphic traits are ubiquitous and play vital roles in reproduction, courtship, and environmental adaptation, especially in insects. In this study, we used the CRISPR/Cas9 genome editing system to generate somatic mutations of the Masculinizer (Masc) and doublesex (dsx) genes in the sex determination pathway of Ostrinia furnacalis. The OfMasc and Ofdsx genes are structural orthologs of the key sex regulation factors in Bombyx mori. Mutation of the OfMasc and Ofdsx genes induced abnormal external genitalia, adult sterility, and sex reversal of sexually dimorphic traits including wing pigmentation, gene expression patterns, and dsx sex-specific splicing. These results demonstrate that the Masc and dsx genes are conserved factors in sexually dimorphic traits, and therefore represent potential target genes in the effort to control O. furnacalis and other lepidopteran pests. Abstract Sex determination is an important and traditional biological process. In Lepidoptera, Masculinizer (Masc) and doublesex (dsx) are the essential genes for sex determination and play critical roles in sexual differentiation and development. The functions of Masc and dsx have been characterized in several model insect species. However, the molecular mechanism and sex determination functions of Masc and dsx in Ostrinia furnacalis, an agricultural pest, are still unknown. Here, we successfully used the CRISPR/Cas9 genome editing system to knock out OfMasc and Ofdsx. Mutation of OfMasc induced male external genital defects and sterility. Disruptions of the Ofdsx common region caused sex-specific defects in the external genitals and adult sterility. In addition, we found that OfMasc and Ofdsx can regulate the pigmentation genes that control wing pigmentation patterns. These results demonstrate that OfMasc and Ofdsx play key roles in the sex determination of O. furnacalis, and suggest novel genetic control approaches for the management of pests, including O. furnacalis.
Collapse
|
6
|
Diao L, Turek PJ, John CM, Fang F, Reijo Pera RA. Roles of Spermatogonial Stem Cells in Spermatogenesis and Fertility Restoration. Front Endocrinol (Lausanne) 2022; 13:895528. [PMID: 35634498 PMCID: PMC9135128 DOI: 10.3389/fendo.2022.895528] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/31/2022] [Indexed: 01/21/2023] Open
Abstract
Spermatogonial stem cells (SSCs) are a group of adult stem cells in the testis that serve as the foundation of continuous spermatogenesis and male fertility. SSCs are capable of self-renewal to maintain the stability of the stem cell pool and differentiation to produce mature spermatozoa. Dysfunction of SSCs leads to male infertility. Therefore, dissection of the regulatory network of SSCs is of great significance in understanding the fundamental molecular mechanisms of spermatogonial stem cell function in spermatogenesis and the pathogenesis of male infertility. Furthermore, a better understanding of SSC biology will allow us to culture and differentiate SSCs in vitro, which may provide novel stem cell-based therapy for assisted reproduction. This review summarizes the latest research progress on the regulation of SSCs, and the potential application of SSCs for fertility restoration through in vivo and in vitro spermatogenesis. We anticipate that the knowledge gained will advance the application of SSCs to improve male fertility. Furthermore, in vitro spermatogenesis from SSCs sets the stage for the production of SSCs from induced pluripotent stem cells (iPSCs) and subsequent spermatogenesis.
Collapse
Affiliation(s)
- Lei Diao
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | | | | | - Fang Fang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Renee A. Reijo Pera
- McLaughlin Research Institute, Touro College of Osteopathic Medicine – Montana (TouroCOM-MT), Great Falls, MT, United States
- Research Division, Touro College of Osteopathic Medicine – Montana (TouroCOM-MT), Great Falls, MT, United States
| |
Collapse
|
7
|
Khan UW, Newmark PA. Somatic regulation of female germ cell regeneration and development in planarians. Cell Rep 2022; 38:110525. [PMID: 35294875 PMCID: PMC8994625 DOI: 10.1016/j.celrep.2022.110525] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/11/2022] [Accepted: 02/22/2022] [Indexed: 12/23/2022] Open
Abstract
Female germ cells develop into oocytes, with the capacity for totipotency. In most animals, these remarkable cells are specified during development and cannot be regenerated. By contrast, planarians, known for their regenerative prowess, can regenerate germ cells. To uncover mechanisms required for female germ cell development and regeneration, we generated gonad-specific transcriptomes and identified genes whose expression defines progressive stages of female germ cell development. Strikingly, early female germ cells share molecular signatures with the pluripotent stem cells driving planarian regeneration. We observe spatial heterogeneity within somatic ovarian cells and find that a regionally enriched foxL homolog is required for oocyte differentiation, but not specification, suggestive of functionally distinct somatic compartments. Unexpectedly, a neurotransmitter-biosynthetic enzyme, aromatic L-amino acid decarboxylase (AADC), is also expressed in somatic gonadal cells, and plays opposing roles in female and male germ cell development. Thus, somatic gonadal cells deploy conserved factors to regulate germ cell development and regeneration in planarians.
Collapse
Affiliation(s)
- Umair W Khan
- Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI, USA; Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Phillip A Newmark
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA; Howard Hughes Medical Institute, Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
8
|
Bronikowski AM, Meisel RP, Biga PR, Walters J, Mank JE, Larschan E, Wilkinson GS, Valenzuela N, Conard AM, de Magalhães JP, Duan J, Elias AE, Gamble T, Graze R, Gribble KE, Kreiling JA, Riddle NC. Sex-specific aging in animals: Perspective and future directions. Aging Cell 2022; 21:e13542. [PMID: 35072344 PMCID: PMC8844111 DOI: 10.1111/acel.13542] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/15/2021] [Accepted: 12/11/2021] [Indexed: 12/14/2022] Open
Abstract
Sex differences in aging occur in many animal species, and they include sex differences in lifespan, in the onset and progression of age-associated decline, and in physiological and molecular markers of aging. Sex differences in aging vary greatly across the animal kingdom. For example, there are species with longer-lived females, species where males live longer, and species lacking sex differences in lifespan. The underlying causes of sex differences in aging remain mostly unknown. Currently, we do not understand the molecular drivers of sex differences in aging, or whether they are related to the accepted hallmarks or pillars of aging or linked to other well-characterized processes. In particular, understanding the role of sex-determination mechanisms and sex differences in aging is relatively understudied. Here, we take a comparative, interdisciplinary approach to explore various hypotheses about how sex differences in aging arise. We discuss genomic, morphological, and environmental differences between the sexes and how these relate to sex differences in aging. Finally, we present some suggestions for future research in this area and provide recommendations for promising experimental designs.
Collapse
Affiliation(s)
- Anne M. Bronikowski
- Department of Ecology, Evolution, and Organismal BiologyIowa State UniversityAmesIowaUSA
| | - Richard P. Meisel
- Department of Biology and BiochemistryUniversity of HoustonHoustonTexasUSA
| | - Peggy R. Biga
- Department of BiologyThe University of Alabama at BirminghamBirminghamAlabamaUSA
| | - James R. Walters
- Department of Ecology and Evolutionary BiologyThe University of KansasLawrenceKansasUSA
| | - Judith E. Mank
- Department of ZoologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of BioscienceUniversity of ExeterPenrynUK
| | - Erica Larschan
- Department of Molecular Biology, Cell Biology and BiochemistryBrown UniversityProvidenceRhode IslandUSA
| | | | - Nicole Valenzuela
- Department of Ecology, Evolution, and Organismal BiologyIowa State UniversityAmesIowaUSA
| | - Ashley Mae Conard
- Department of Computer ScienceCenter for Computational and Molecular BiologyBrown UniversityProvidenceRhode IslandUSA
| | - João Pedro de Magalhães
- Integrative Genomics of Ageing GroupInstitute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
| | | | - Amy E. Elias
- Department of Molecular Biology, Cell Biology and BiochemistryBrown UniversityProvidenceRhode IslandUSA
| | - Tony Gamble
- Department of Biological SciencesMarquette UniversityMilwaukeeWisconsinUSA
- Milwaukee Public MuseumMilwaukeeWisconsinUSA
- Bell Museum of Natural HistoryUniversity of MinnesotaSaint PaulMinnesotaUSA
| | - Rita M. Graze
- Department of Biological SciencesAuburn UniversityAuburnAlabamaUSA
| | - Kristin E. Gribble
- Josephine Bay Paul Center for Comparative Molecular Biology and EvolutionMarine Biological LaboratoryWoods HoleMassachusettsUSA
| | - Jill A. Kreiling
- Department of Molecular Biology, Cell Biology and BiochemistryBrown UniversityProvidenceRhode IslandUSA
| | - Nicole C. Riddle
- Department of BiologyThe University of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
9
|
Reilein A, Kogan HV, Misner R, Park KS, Kalderon D. Adult stem cells and niche cells segregate gradually from common precursors that build the adult Drosophila ovary during pupal development. eLife 2021; 10:69749. [PMID: 34590579 PMCID: PMC8536258 DOI: 10.7554/elife.69749] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 09/29/2021] [Indexed: 12/31/2022] Open
Abstract
Production of proliferative follicle cells (FCs) and quiescent escort cells (ECs) by follicle stem cells (FSCs) in adult Drosophila ovaries is regulated by niche signals from anterior (cap cells, ECs) and posterior (polar FCs) sources. Here we show that ECs, FSCs, and FCs develop from common pupal precursors, with different fates acquired by progressive separation of cells along the AP axis and a graded decline in anterior cell proliferation. ECs, FSCs, and most FCs derive from intermingled cell (IC) precursors interspersed with germline cells. Precursors also accumulate posterior to ICs before engulfing a naked germline cyst projected out of the germarium to form the first egg chamber and posterior polar FC signaling center. Thus, stem and niche cells develop in appropriate numbers and spatial organization through regulated proliferative expansion together with progressive establishment of spatial signaling cues that guide adult cell behavior, rather than through rigid early specification events.
Collapse
Affiliation(s)
- Amy Reilein
- Department of Biological Sciences, Columbia University, New York, United States
| | - Helen V Kogan
- Department of Biological Sciences, Columbia University, New York, United States
| | - Rachel Misner
- Department of Biological Sciences, Columbia University, New York, United States
| | - Karen Sophia Park
- Department of Biological Sciences, Columbia University, New York, United States
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, United States
| |
Collapse
|
10
|
Mine S, Sumitani M, Aoki F, Hatakeyama M, Suzuki MG. Effects of Functional Depletion of Doublesex on Male Development in the Sawfly, Athalia rosae. INSECTS 2021; 12:insects12100849. [PMID: 34680618 PMCID: PMC8538284 DOI: 10.3390/insects12100849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 01/04/2023]
Abstract
Simple Summary The sawfly, Athalia rosae, exploits a haplodiploid mode of reproduction, in which fertilized eggs develop into diploid females, whereas unfertilized eggs parthenogenetically develop into haploid males. The doublesex (dsx) gene is a well-conserved transcription factor that regulates sexual differentiation in insects. In the present study, we knocked down the A. rosae ortholog of dsx (Ardsx) during several developmental stages with repeated double-stranded RNA (dsRNA) injections. As a result, knockdown of Ardsx in haploid males caused almost complete male-to-female sex reversal, but the resulting eggs were infertile. The same knockdown approach using diploid males caused complete male-to-female sex reversal; they were able to produce fertile eggs and exhibited female behaviors. The same RNAi treatment did not affect female differentiation. These results demonstrated that dsx in the sawfly is essential for male development and its depletion caused complete male-to-female sex reversal. This is the first demonstration of functional depletion of dsx not causing intersexuality but inducing total sex reversal in males instead. Abstract The doublesex (dsx) gene, which encodes a transcription factor, regulates sexual differentiation in insects. Sex-specific splicing of dsx occurs to yield male- and female-specific isoforms, which promote male and female development, respectively. Thus, functional disruption of dsx leads to an intersexual phenotype in both sexes. We previously identified a dsx ortholog in the sawfly, Athalia rosae. Similar to dsx in other insects, dsx in the sawfly yields different isoforms in males and females as a result of alternative splicing. The sawfly exploits a haplodiploid mode of reproduction, in which fertilized eggs develop into diploid females, whereas unfertilized eggs parthenogenetically develop into haploid males. In the present study, we knocked down the A. rosae ortholog of dsx (Ardsx) during several developmental stages with repeated double-stranded RNA (dsRNA) injections. Knockdown of Ardsx via parental RNA interference (RNAi), which enables knockdown of genes in offspring embryos, led to a lack of internal and external genitalia in haploid male progeny. Additional injection of dsRNA targeting Ardsx in these animals caused almost complete male-to-female sex reversal, but the resulting eggs were infertile. Notably, the same knockdown approach using diploid males obtained by sib-crossing caused complete male-to-female sex reversal; they were morphologically and behaviorally females. The same RNAi treatment did not affect female differentiation. These results indicate that dsx in the sawfly is essential for male development and its depletion caused complete male-to-female sex reversal. This is the first demonstration of functional depletion of dsx not causing intersexuality but inducing total sex reversal in males instead.
Collapse
Affiliation(s)
- Shotaro Mine
- Department of Biosciences, Nihon University, 3-25-40 Sakurajosui, Setagaya-ku, Tokyo 156-8550, Japan;
| | - Megumi Sumitani
- Division of Biotechnology, Institute of Agrobiological Sciences, NARO, Owashi, Tsukuba 305-8634, Japan;
| | - Fugaku Aoki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8562, Japan;
| | - Masatsugu Hatakeyama
- Division of Applied Genetics, Institute of Agrobiological Sciences, NARO, Owashi, Tsukuba 305-8634, Japan;
| | - Masataka G. Suzuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa 277-8562, Japan;
- Correspondence: ; Tel.: +81-4-7136-3694
| |
Collapse
|
11
|
Wang Y, Xu X, Chen X, Li X, Bi H, Xu J, Zhu C, Niu C, Huang Y. Mutation of P-element somatic inhibitor induces male sterility in the diamondback moth, Plutella xylostella. PEST MANAGEMENT SCIENCE 2021; 77:3588-3596. [PMID: 33843144 DOI: 10.1002/ps.6413] [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: 10/23/2020] [Revised: 03/29/2021] [Accepted: 04/11/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Genetic manipulation of sex determination pathways in insects provides the basis for a broad range of strategies to benefit agricultural security and human health. The P-element somatic inhibitor (PSI) protein, an exon splicing silencer that promotes male-specific splicing of dsx, plays a critical role in male sexual differentiation and development. The functions of PSI have been characterized in the lepidopteran model species Bombyx mori. However, the molecular mechanism and functions of PSI in Plutella xylostella, a worldwide agricultural pest and taxonomically basal species, are still unknown. RESULTS Here we identified PxPSI transcripts and analyzed their spatiotemporal expression pattern in P. xylostella. Multiple sequence alignment revealed that PxPSI contains four KH domains and is highly conserved in lepidopterans. We used the CRISPR-Cas9 system to generate mutations of the PxPSI genomic locus. Disruptions of PxPSI caused male-specific defects in internal and external genitals. In addition, we detected female-specific Pxdsx transcripts in PxPSI male mutants. Mutations also caused changes in expression of several sex-biased genes and induced male sterility. CONCLUSION Our study demonstrates that PxPSI plays a key role in male sex determination in P. xylostella and suggests a potential molecular target for genetic-based pest management in lepidopteran pests. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yaohui Wang
- Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, College of Plant Science & Technology, Huazhong Agricultural University, Wuhan, China
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences/Institute of Plant Physiology and Ecology, Shanghai, China
| | - Xia Xu
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences/Institute of Plant Physiology and Ecology, Shanghai, China
| | - Xi'en Chen
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences/Institute of Plant Physiology and Ecology, Shanghai, China
| | - Xiaowei Li
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences/Institute of Plant Physiology and Ecology, Shanghai, China
| | - Honglun Bi
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences/Institute of Plant Physiology and Ecology, Shanghai, China
| | - Jun Xu
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences/Institute of Plant Physiology and Ecology, Shanghai, China
| | - Chenxu Zhu
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences/Institute of Plant Physiology and Ecology, Shanghai, China
| | - Changying Niu
- Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, College of Plant Science & Technology, Huazhong Agricultural University, Wuhan, China
| | - Yongping Huang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences/Institute of Plant Physiology and Ecology, Shanghai, China
| |
Collapse
|
12
|
Li YR, Lai HW, Huang HH, Chen HC, Fugmann SD, Yang SY. Trajectory mapping of the early Drosophila germline reveals controls of zygotic activation and sex differentiation. Genome Res 2021; 31:1011-1023. [PMID: 33858841 PMCID: PMC8168578 DOI: 10.1101/gr.271148.120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 04/07/2021] [Indexed: 01/29/2023]
Abstract
Germ cells in Drosophila melanogaster are specified maternally shortly after fertilization and are transcriptionally quiescent until their zygotic genome is activated to sustain further development. To understand the molecular basis of this process, we analyzed the progressing transcriptomes of early male and female germ cells at the single-cell level between germline specification and coalescence with somatic gonadal cells. Our data comprehensively cover zygotic activation in the germline genome, and analyses on genes that exhibit germline-restricted expression reveal that polymerase pausing and differential RNA stability are important mechanisms that establish gene expression differences between the germline and soma. In addition, we observe an immediate bifurcation between the male and female germ cells as zygotic transcription begins. The main difference between the two sexes is an elevation in X Chromosome expression in females relative to males, signifying incomplete dosage compensation, with a few select genes exhibiting even higher expression increases. These indicate that the male program is the default mode in the germline that is driven to female development with a second X Chromosome.
Collapse
Affiliation(s)
- Yi-Ru Li
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan
| | - Hsiao Wen Lai
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan
| | - Hsiao Han Huang
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan
| | - Hsing-Chun Chen
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan
| | - Sebastian D Fugmann
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan.,Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan.,Department of Nephrology, Linkou Chang Gung Memorial Hospital, Kweishan, Taoyuan 333 Taiwan
| | - Shu Yuan Yang
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan.,Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan.,Department of Gynecology, Linkou Chang Gung Memorial Hospital, Kweishan, Taoyuan 333 Taiwan
| |
Collapse
|
13
|
Absence of X-chromosome dosage compensation in the primordial germ cells of Drosophila embryos. Sci Rep 2021; 11:4890. [PMID: 33649478 PMCID: PMC7921590 DOI: 10.1038/s41598-021-84402-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/16/2021] [Indexed: 01/31/2023] Open
Abstract
Dosage compensation is a mechanism that equalizes sex chromosome gene expression between the sexes. In Drosophila, individuals with two X chromosomes (XX) become female, whereas males have one X chromosome (XY). In males, dosage compensation of the X chromosome in the soma is achieved by five proteins and two non-coding RNAs, which assemble into the male-specific lethal (MSL) complex to upregulate X-linked genes twofold. By contrast, it remains unclear whether dosage compensation occurs in the germline. To address this issue, we performed transcriptome analysis of male and female primordial germ cells (PGCs). We found that the expression levels of X-linked genes were approximately twofold higher in female PGCs than in male PGCs. Acetylation of lysine residue 16 on histone H4 (H4K16ac), which is catalyzed by the MSL complex, was undetectable in these cells. In male PGCs, hyperactivation of X-linked genes and H4K16ac were induced by overexpression of the essential components of the MSL complex, which were expressed at very low levels in PGCs. Together, these findings indicate that failure of MSL complex formation results in the absence of X-chromosome dosage compensation in male PGCs.
Collapse
|
14
|
Yuzawa T, Matsuoka M, Sumitani M, Aoki F, Sezutsu H, Suzuki MG. Transgenic and knockout analyses of Masculinizer and doublesex illuminated the unique functions of doublesex in germ cell sexual development of the silkworm, Bombyx mori. BMC DEVELOPMENTAL BIOLOGY 2020; 20:19. [PMID: 32957956 PMCID: PMC7504827 DOI: 10.1186/s12861-020-00224-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/10/2020] [Indexed: 01/24/2023]
Abstract
Background Masculinizer (Masc) plays a pivotal role in male sex determination in the silkworm, Bombyx mori. Masc is required for male-specific splicing of B. mori doublesex (Bmdsx) transcripts. The male isoform of Bmdsx (BmdsxM) induces male differentiation in somatic cells, while females express the female isoform of Bmdsx (BmdsxF), which promotes female differentiation in somatic cells. Our previous findings suggest that Masc could direct the differentiation of genetically female (ZW) germ cells into sperms. However, it remains unclear whether Masc directly induces spermatogenesis or if it promotes male differentiation in germ cells indirectly by inducing the expression of BmdsxM. Results In this study, we performed genetic analyses using the transgenic line that expressed Masc, as well as various Bmdsx knockout lines. We found that Masc-expressing females with a homozygous mutation in BmdsxM showed normal development in ovaries. The formation of testis-like tissues was abolished in these females. On the other hand, Masc-expressing females carrying a homozygous mutation in BmdsxF exhibited almost complete male-specific development in gonads and germ cells. These results suggest that BmdsxM has an ability to induce male development in germ cells as well as internal genital organs, while BmdsxF inhibits BmdsxM activity and represses male differentiation. To investigate whether MASC directly controls male-specific splicing of Bmdsx and identify RNAs that form complexes with MASC in testes, we performed RNA immunoprecipitation (RIP) using an anti-MASC antibody. We found that MASC formed a complex with AS1 lncRNA, which is a testis-specific factor involved in the male-specific splicing of Bmdsx pre-mRNA. Conclusions Taken together, our findings suggest that Masc induces male differentiation in germ cells by enhancing the production of BmdsxM. Physical interaction between MASC and AS1 lncRNA may be important for the BmdsxM expression in the testis. Unlike in the Drosophila dsx, BmdsxM was able to induce spermatogenesis in genetically female (ZW) germ cells. To the best of our knowledge, this is the first report that the role of dsx in germ cell sexual development is different between insect species.
Collapse
Affiliation(s)
- Tomohisa Yuzawa
- AIR WATER INC, 4-9-4 Hatchobori, Chuo-ku, Tokyo, 104-0032, Japan.,Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba, 277-8562, Japan
| | - Misato Matsuoka
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba, 277-8562, Japan.,SHINYUSHA, 1-12 Kanda Jimbocho, Chiyoda-ku, Tokyo, 101-0051, Japan
| | - Megumi Sumitani
- Genetically Modified Organism Research Center, National Institute of Agrobiological Sciences, Owashi, Tsukuba, 305-8634, Japan
| | - Fugaku Aoki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba, 277-8562, Japan
| | - Hideki Sezutsu
- Genetically Modified Organism Research Center, National Institute of Agrobiological Sciences, Owashi, Tsukuba, 305-8634, Japan
| | - Masataka G Suzuki
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa-shi, Chiba, 277-8562, Japan.
| |
Collapse
|
15
|
Xu X, Yang J, Harvey-Samuel T, Huang Y, Asad M, Chen W, He W, Yang G, Alphey L, You M. Identification and characterization of the vasa gene in the diamondback moth, Plutella xylostella. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 122:103371. [PMID: 32283279 DOI: 10.1016/j.ibmb.2020.103371] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 06/11/2023]
Abstract
Vasa is an ATP-dependent RNA helicase, participating in multiple biological processes. It has been widely used as a germ cell marker and its promoter has become a key component of several genetic pest control systems. Here we present the vasa gene structure and its promoter activity in Plutella xylostella, one of the most destructive pests of cruciferous crops. Full length Pxvasa cDNA sequences were obtained, revealing 14 exons and at least 30 alternatively spliced transcripts. Inferred amino acid sequences showed nine conserved DEAD-box family protein motifs with partial exclusion from some isoforms. Real-time quantitative PCR indicated the up-regulation of Pxvasa in both female and male adults compared with other developmental stages, and the expression levels of Pxvasa were found to be much higher in adult gonads, especially ovaries, than in other tissues. The putative promoter region of Pxvasa was sequenced and several ecdysone-induced transcription factor (TF) binding sites were predicted in silico. To further analyze the promoter region, two upstream regulatory fragments of different lengths were tested as putative promoters in transient cell and embryo expression assays, one of which was subsequently utilized to drive Cas9 expression in vivo. A transgenic line was recovered and the expression patterns of Cas9 and native Pxvasa were profiled in adult tissues and eggs with RT-PCR. This work provides the foundation for further studies on the gene functions of Pxvasa as well as the potential application of its promoter in genetic manipulation of P. xylostella.
Collapse
Affiliation(s)
- Xuejiao Xu
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture, Fuzhou, 350002, China
| | - Jie Yang
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture, Fuzhou, 350002, China
| | - Tim Harvey-Samuel
- Arthropod Genetics Group, The Pirbright Institute, Woking, GU24 0NF, UK
| | - Yuping Huang
- Department of Physiology & Neurobiology, University of Connecticut, Storrs, CT, 06269, USA
| | - Muhammad Asad
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture, Fuzhou, 350002, China
| | - Wei Chen
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture, Fuzhou, 350002, China
| | - Weiyi He
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture, Fuzhou, 350002, China
| | - Guang Yang
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture, Fuzhou, 350002, China
| | - Luke Alphey
- Arthropod Genetics Group, The Pirbright Institute, Woking, GU24 0NF, UK
| | - Minsheng You
- State Key Laboratory for Ecological Pest Control of Fujian and Taiwan Crops, Institute of Applied Ecology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou 350002, China; Key Laboratory of Integrated Pest Management for Fujian-Taiwan Crops, Ministry of Agriculture, Fuzhou, 350002, China.
| |
Collapse
|
16
|
Xu J, Liu W, Yang D, Chen S, Chen K, Liu Z, Yang X, Meng J, Zhu G, Dong S, Zhang Y, Zhan S, Wang G, Huang Y. Regulation of olfactory-based sex behaviors in the silkworm by genes in the sex-determination cascade. PLoS Genet 2020; 16:e1008622. [PMID: 32520935 PMCID: PMC7307793 DOI: 10.1371/journal.pgen.1008622] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/22/2020] [Accepted: 05/04/2020] [Indexed: 11/28/2022] Open
Abstract
Insect courtship and mating depend on integration of olfactory, visual, and tactile cues. Compared to other insects, Bombyx mori, the domesticated silkworm, has relatively simple sexual behaviors as it cannot fly. Here by using CRISPR/Cas9 and electrophysiological techniques we found that courtship and mating behaviors are regulated in male silk moths by mutating genes in the sex determination cascade belonging to two conserved pathways. Loss of Bmdsx gene expression significantly reduced the peripheral perception of the major pheromone component bombykol by reducing expression of the product of the BmOR1 gene which completely blocked courtship in adult males. Interestingly, we found that mating behavior was regulated independently by another sexual differentiation gene, Bmfru. Loss of Bmfru completely blocked mating, but males displayed normal courtship behavior. Lack of Bmfru expression significantly reduced the perception of the minor pheromone component bombykal due to the down regulation of BmOR3 expression; further, functional analysis revealed that loss of the product of BmOR3 played a key role in terminating male mating behavior. Our results suggest that Bmdsx and Bmfru are at the base of the two primary pathways that regulate olfactory-based sexual behavior. The fundamental insect sexual behaviors, courtship and mating, result from successful integration of olfactory, vision, tactile and other complex innate behaviors. In the widely used insect model, Drosophila melanogaster, the sex determination cascade genes fruitless and doublesex are involved in the regulation of courtship and mating behaviors; however, little is known about the function of these sexual differentiation genes in regulating sex behaviors of Lepidoptera. Here we combine genetics and electrophysiology to investigate regulation pathway of sexual behaviors in the model lepidopteran insect, the domesticated silk moth, Bombyx mori. Our results support the presence of two genetic pathways in B. mori, named Bmdsx-BmOR1-bombykol and Bmfru-BmOR3-bombykal, which control distinct aspects of male sexual behavior that are modulated by olfaction. This is the first comprehensive report about the role of sex differentiation genes in the male sexual behavior in the silk moth.
Collapse
Affiliation(s)
- Jun Xu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Wei Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Dehong Yang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Shuqing Chen
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Kai Chen
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Zulian Liu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Xu Yang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Jing Meng
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guanheng Zhu
- Education Ministry Key Laboratory of Integrated Management of Crop Disease and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Shuanglin Dong
- Education Ministry Key Laboratory of Integrated Management of Crop Disease and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Yong Zhang
- Department of Biology, University of Nevada, Reno, Nevada, United States of America
| | - Shuai Zhan
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Guirong Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail: (GW); (YH)
| | - Yongping Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (GW); (YH)
| |
Collapse
|
17
|
Barreñada O, Fernández-Pérez D, Larriba E, Brieño-Enriquez M, Del Mazo J. Diversification of piRNAs expressed in PGCs and somatic cells during embryonic gonadal development. RNA Biol 2020; 17:1309-1323. [PMID: 32375541 DOI: 10.1080/15476286.2020.1757908] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
piRNAs are small non-coding RNAs known to play a main role in defence against transposable elements in germ cells. However, other potential functions, such as biogenesis and differences in somatic and germline expression of these regulatory elements, are not yet fully unravelled. Here, we analysed a variety of piRNA sequences detected in mouse male and female primordial germ cells (PGCs) and gonadal somatic cells at crucial stages during embryonic differentiation of germ cells (11.5-13.5 days post-coitum). NGS of sncRNA and bioinformatic characterization of piRNAs from PGCs and somatic cells, in addition to piRNAs associated with TEs, indicated functional diversification in both cell types. Differences in the proportion of the diverse types of piRNAs are detected between somatic and germline during development. However, the global diversified patterns of piRNA expression are mainly shared between germ and somatic cells, we identified piRNAs related with molecules involved in ribosome components and translation pathway, including piRNAs derived from rRNA (34%), tRNA (10%) and snoRNA (8%). piRNAs from both tRNA and snoRNA are mainly derived from 3' and 5' end regions. These connections between piRNAs and rRNAs, tRNAs or snoRNAs suggest important functions of specialized piRNAs in translation regulation during this window of gonadal development.
Collapse
Affiliation(s)
- Odei Barreñada
- Department of Cellular & Molecular Biology, Centro De Investigaciones Biológicas C.I.B. (CSIC) , Madrid, Spain
| | - Daniel Fernández-Pérez
- Department of Cellular & Molecular Biology, Centro De Investigaciones Biológicas C.I.B. (CSIC) , Madrid, Spain
| | - Eduardo Larriba
- Department of Cellular & Molecular Biology, Centro De Investigaciones Biológicas C.I.B. (CSIC) , Madrid, Spain
| | - Miguel Brieño-Enriquez
- Department of Cellular & Molecular Biology, Centro De Investigaciones Biológicas C.I.B. (CSIC) , Madrid, Spain
| | - Jesús Del Mazo
- Department of Cellular & Molecular Biology, Centro De Investigaciones Biológicas C.I.B. (CSIC) , Madrid, Spain
| |
Collapse
|
18
|
Aoki ST, Porter DF, Prasad A, Wickens M, Bingman CA, Kimble J. An RNA-Binding Multimer Specifies Nematode Sperm Fate. Cell Rep 2019; 23:3769-3775. [PMID: 29949762 PMCID: PMC6066285 DOI: 10.1016/j.celrep.2018.05.095] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/26/2018] [Accepted: 05/30/2018] [Indexed: 11/27/2022] Open
Abstract
FOG-3 is a master regulator of sperm fate in Caenorhabditis elegans and homologous to Tob/BTG proteins, which in mammals are monomeric adaptors that recruit enzymes to RNA binding proteins. Here, we determine the FOG-3 crystal structure and in vitro demonstrate that FOG-3 forms dimers that can multi-merize. The FOG-3 multimeric structure has a basic surface potential, suggestive of binding nucleic acid. Consistent with that prediction, FOG-3 binds directly to nearly 1,000 RNAs in nematode spermatogenic germ cells. Most binding is to the 3′ UTR, and most targets (94%) are oogenic mRNAs, even though assayed in spermatogenic cells. When tethered to a reporter mRNA, FOG-3 represses its expression. Together these findings elucidate the molecular mechanism of sperm fate specification and reveal the evolution of a protein from monomeric to multimeric form with acquisition of a distinct mode of mRNA repression. The mechanism of the sperm or oocyte fate decision has been elusive. Aoki et al. report that nematode FOG-3, a Tob/BTG protein driving sperm fate, has evolved from monomeric to multimeric form with acquisition of a divergent Tob/BTG mechanism for mRNA repression.
Collapse
Affiliation(s)
- Scott T Aoki
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Douglas F Porter
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Aman Prasad
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Marvin Wickens
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Craig A Bingman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Judith Kimble
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
19
|
Benner L, Castro EA, Whitworth C, Venken KJT, Yang H, Fang J, Oliver B, Cook KR, Lerit DA. Drosophila Heterochromatin Stabilization Requires the Zinc-Finger Protein Small Ovary. Genetics 2019; 213:877-895. [PMID: 31558581 PMCID: PMC6827387 DOI: 10.1534/genetics.119.302590] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/21/2019] [Indexed: 02/04/2023] Open
Abstract
Heterochromatin-mediated repression is essential for controlling the expression of transposons and for coordinated cell type-specific gene regulation. The small ovary (sov) locus was identified in a screen for female-sterile mutations in Drosophila melanogaster, and mutants show dramatic ovarian morphogenesis defects. We show that the null sov phenotype is lethal and map the locus to the uncharacterized gene CG14438, which encodes a nuclear zinc-finger protein that colocalizes with the essential Heterochromatin Protein 1 (HP1a). We demonstrate Sov functions to repress inappropriate gene expression in the ovary, silence transposons, and suppress position-effect variegation in the eye, suggesting a central role in heterochromatin stabilization.
Collapse
Affiliation(s)
- Leif Benner
- Section of Developmental Genomics, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
- Department of Biology, Johns Hopkins University, Baltimore, Maryland 21218
| | - Elias A Castro
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Cale Whitworth
- Section of Developmental Genomics, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
- Department of Biology, Indiana University, Bloomington, Indiana 47405
| | - Koen J T Venken
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology
- McNair Medical Institute at the Robert and Janice McNair Foundation
- Dan L. Duncan Cancer Center, Center for Drug Discovery
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas 77030
| | - Haiwang Yang
- Section of Developmental Genomics, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Junnan Fang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322
| | - Brian Oliver
- Section of Developmental Genomics, Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892
| | - Kevin R Cook
- Department of Biology, Indiana University, Bloomington, Indiana 47405
| | - Dorothy A Lerit
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
20
|
Tudor-domain containing protein 5-like promotes male sexual identity in the Drosophila germline and is repressed in females by Sex lethal. PLoS Genet 2019; 15:e1007617. [PMID: 31329582 PMCID: PMC6645463 DOI: 10.1371/journal.pgen.1007617] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 06/26/2019] [Indexed: 11/19/2022] Open
Abstract
For sexually reproducing organisms, production of male or female gametes depends on specifying the correct sexual identity in the germline. In D. melanogaster, Sex lethal (Sxl) is the key gene that controls sex determination in both the soma and the germline, but how it does so in the germline is unknown, other than that it is different than in the soma. We conducted an RNA expression profiling experiment to identify direct and indirect germline targets of Sxl specifically in the undifferentiated germline. We find that, in these cells, Sxl loss does not lead to a global masculinization observed at the whole-genome level. In contrast, Sxl appears to affect a discrete set of genes required in the male germline, such as Phf7. We also identify Tudor domain containing protein 5-like (Tdrd5l) as a target for Sxl regulation that is important for male germline identity. Tdrd5l is repressed by Sxl in female germ cells, but is highly expressed in male germ cells where it promotes proper male fertility and germline differentiation. Additionally, Tdrd5l localizes to cytoplasmic granules with some characteristics of RNA Processing (P-) Bodies, suggesting that it promotes male identity in the germline by regulating post-transcriptional gene expression.
Collapse
|
21
|
Primus S, Pozmanter C, Baxter K, Van Doren M. Tudor-domain containing protein 5-like promotes male sexual identity in the Drosophila germline and is repressed in females by Sex lethal. PLoS Genet 2019. [PMID: 31329582 DOI: 10.1101/388850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023] Open
Abstract
For sexually reproducing organisms, production of male or female gametes depends on specifying the correct sexual identity in the germline. In D. melanogaster, Sex lethal (Sxl) is the key gene that controls sex determination in both the soma and the germline, but how it does so in the germline is unknown, other than that it is different than in the soma. We conducted an RNA expression profiling experiment to identify direct and indirect germline targets of Sxl specifically in the undifferentiated germline. We find that, in these cells, Sxl loss does not lead to a global masculinization observed at the whole-genome level. In contrast, Sxl appears to affect a discrete set of genes required in the male germline, such as Phf7. We also identify Tudor domain containing protein 5-like (Tdrd5l) as a target for Sxl regulation that is important for male germline identity. Tdrd5l is repressed by Sxl in female germ cells, but is highly expressed in male germ cells where it promotes proper male fertility and germline differentiation. Additionally, Tdrd5l localizes to cytoplasmic granules with some characteristics of RNA Processing (P-) Bodies, suggesting that it promotes male identity in the germline by regulating post-transcriptional gene expression.
Collapse
Affiliation(s)
- Shekerah Primus
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Caitlin Pozmanter
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Kelly Baxter
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Mark Van Doren
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, United States of America
| |
Collapse
|
22
|
Millington JW, Rideout EJ. Sex differences in Drosophila development and physiology. CURRENT OPINION IN PHYSIOLOGY 2018. [DOI: 10.1016/j.cophys.2018.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
23
|
The H3K9 methyltransferase SETDB1 maintains female identity in Drosophila germ cells. Nat Commun 2018; 9:4155. [PMID: 30297796 PMCID: PMC6175928 DOI: 10.1038/s41467-018-06697-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/19/2018] [Indexed: 12/17/2022] Open
Abstract
The preservation of germ cell sexual identity is essential for gametogenesis. Here we show that H3K9me3-mediated gene silencing is integral to female fate maintenance in Drosophila germ cells. Germ cell specific loss of the H3K9me3 pathway members, the H3K9 methyltransferase SETDB1, WDE, and HP1a, leads to ectopic expression of genes, many of which are normally expressed in testis. SETDB1 controls the accumulation of H3K9me3 over a subset of these genes without spreading into neighboring loci. At phf7, a regulator of male germ cell sexual fate, the H3K9me3 peak falls over the silenced testis-specific transcription start site. Furthermore, H3K9me3 recruitment to phf7 and repression of testis-specific transcription is dependent on the female sex determination gene Sxl. Thus, female identity is secured by an H3K9me3 epigenetic pathway in which Sxl is the upstream female-specific regulator, SETDB1 is the required chromatin writer, and phf7 is one of the critical SETDB1 target genes. Epigenetic regulation is critical for the maintenance of germ cell identity. Here the authors show that H3K9me3-mediated gene silencing is critical for repression of testis-specific transcription in Drosophila female germ cells, indicating H3K9me3 maintains female germ cell sexual identity.
Collapse
|
24
|
Kyrou K, Hammond AM, Galizi R, Kranjc N, Burt A, Beaghton AK, Nolan T, Crisanti A. A CRISPR-Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes. Nat Biotechnol 2018; 36:1062-1066. [PMID: 30247490 PMCID: PMC6871539 DOI: 10.1038/nbt.4245] [Citation(s) in RCA: 487] [Impact Index Per Article: 69.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 08/03/2018] [Indexed: 01/13/2023]
Abstract
Complete population collapse of malaria vector Anopheles gambiae in cages is achieved using a gene drive that targets doublesex. In the human malaria vector Anopheles gambiae, the gene doublesex (Agdsx) encodes two alternatively spliced transcripts, dsx-female (AgdsxF) and dsx-male (AgdsxM), that control differentiation of the two sexes. The female transcript, unlike the male, contains an exon (exon 5) whose sequence is highly conserved in all Anopheles mosquitoes so far analyzed. We found that CRISPR–Cas9-targeted disruption of the intron 4–exon 5 boundary aimed at blocking the formation of functional AgdsxF did not affect male development or fertility, whereas females homozygous for the disrupted allele showed an intersex phenotype and complete sterility. A CRISPR–Cas9 gene drive construct targeting this same sequence spread rapidly in caged mosquitoes, reaching 100% prevalence within 7–11 generations while progressively reducing egg production to the point of total population collapse. Owing to functional constraint of the target sequence, no selection of alleles resistant to the gene drive occurred in these laboratory experiments. Cas9-resistant variants arose in each generation at the target site but did not block the spread of the drive.
Collapse
Affiliation(s)
- Kyros Kyrou
- Department of Life Sciences, Imperial College London, UK
| | | | - Roberto Galizi
- Department of Life Sciences, Imperial College London, UK
| | - Nace Kranjc
- Department of Life Sciences, Imperial College London, UK
| | - Austin Burt
- Department of Life Sciences, Imperial College London, UK
| | | | - Tony Nolan
- Department of Life Sciences, Imperial College London, UK
| | | |
Collapse
|
25
|
Zhang W, Bi Y, Wang Y, Wang M, Li D, Cheng S, Jin J, Li T, Li B, Zhang Y. Nanos2 promotes differentiation of male germ cells basing on the negative regulation of Foxd3 and the treatment of 5-Azadc and TSA. J Cell Physiol 2018; 234:3762-3774. [PMID: 30146792 DOI: 10.1002/jcp.27139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/09/2018] [Indexed: 11/07/2022]
Abstract
The transcription factor positioning in promoter regions relate to gene regulation, and the level of DNA methylation and histone acetylation also impact the promoter activity. In this study, we tested and verified the core promoter region and key transcription factor of Nanos2 which is a male-critical gene in the differentiation of embryonic stem cells to male germ cells, meanwhile, epigenetic effects by mean of 5-Aza-2'-deoxycytidine (5-Azadc) and Trichostin A (TSA) on the activity of Nanos2 promoter were detected. The results reveal that key transcription factor Foxd3 is a negative regulator of Nanos2, which suggests that loss-of-function of Foxd3 causes strong expression of Nanos2 responsive to large amounts of primordial germ cells and spermatogonial stem cells,whereas its overexpression causes the opposite effect. Furthermore, both 5-Azadc and TSA can provoke responses of Nanos2, but the combination effect of the two is better.
Collapse
Affiliation(s)
- Wenhui Zhang
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| | - Yulin Bi
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| | - Yingjie Wang
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| | - Man Wang
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| | - Dong Li
- Reproductive Medicine Center, Drum Tower Clinic Medical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shaoze Cheng
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| | - Jing Jin
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| | - Tingting Li
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| | - Bichun Li
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| | - Yani Zhang
- College of Animal Science and Technology, Yangzhou University, Jiangsu Province Key Laboratory of Animal Breeding and Molecular Design, Yangzhou, Jiangsu, China
| |
Collapse
|
26
|
Grmai L, Hudry B, Miguel-Aliaga I, Bach EA. Chinmo prevents transformer alternative splicing to maintain male sex identity. PLoS Genet 2018; 14:e1007203. [PMID: 29389999 PMCID: PMC5811060 DOI: 10.1371/journal.pgen.1007203] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 02/13/2018] [Accepted: 01/16/2018] [Indexed: 01/15/2023] Open
Abstract
Reproduction in sexually dimorphic animals relies on successful gamete production, executed by the germline and aided by somatic support cells. Somatic sex identity in Drosophila is instructed by sex-specific isoforms of the DMRT1 ortholog Doublesex (Dsx). Female-specific expression of Sex-lethal (Sxl) causes alternative splicing of transformer (tra) to the female isoform traF. In turn, TraF alternatively splices dsx to the female isoform dsxF. Loss of the transcriptional repressor Chinmo in male somatic stem cells (CySCs) of the testis causes them to "feminize", resembling female somatic stem cells in the ovary. This somatic sex transformation causes a collapse of germline differentiation and male infertility. We demonstrate this feminization occurs by transcriptional and post-transcriptional regulation of traF. We find that chinmo-deficient CySCs upregulate tra mRNA as well as transcripts encoding tra-splice factors Virilizer (Vir) and Female lethal (2)d (Fl(2)d). traF splicing in chinmo-deficient CySCs leads to the production of DsxF at the expense of the male isoform DsxM, and both TraF and DsxF are required for CySC sex transformation. Surprisingly, CySC feminization upon loss of chinmo does not require Sxl but does require Vir and Fl(2)d. Consistent with this, we show that both Vir and Fl(2)d are required for tra alternative splicing in the female somatic gonad. Our work reveals the need for transcriptional regulation of tra in adult male stem cells and highlights a previously unobserved Sxl-independent mechanism of traF production in vivo. In sum, transcriptional control of the sex determination hierarchy by Chinmo is critical for sex maintenance in sexually dimorphic tissues and is vital in the preservation of fertility.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Bruno Hudry
- MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| | - Irene Miguel-Aliaga
- MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| | - Erika A. Bach
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Kimmel Stem Cell Center, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
27
|
Ota R, Morita S, Sato M, Shigenobu S, Hayashi M, Kobayashi S. Transcripts immunoprecipitated with Sxl protein in primordial germ cells of Drosophila embryos. Dev Growth Differ 2017; 59:713-723. [PMID: 29124738 DOI: 10.1111/dgd.12408] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/26/2017] [Accepted: 09/27/2017] [Indexed: 12/11/2022]
Abstract
In Drosophila, Sex lethal (Sxl), an RNA binding protein, is required for induction of female sexual identity in both somatic and germline cells. Although the Sxl-dependent feminizing pathway in the soma was previously elucidated, the downstream targets for Sxl in the germline remained elusive. To identify these target genes, we selected transcripts associated with Sxl in primordial germ cells (PGCs) of embryos using RNA immunoprecipitation coupled to sequencing (RIP-seq) analysis. A total of 308 transcripts encoded by 282 genes were obtained. Seven of these genes, expressed at higher levels in PGCs as determined by microarray and in situ hybridization analyses, were subjected to RNAi-mediated functional analyses. Knockdown of Neos, Kap-alpha3, and CG32075 throughout germline development caused gonadal dysgenesis in a sex-dependent manner, and Su(var)2-10 knockdown caused gonadal dysgenesis in both sexes. Moreover, as with knockdown of Sxl, knockdown of Su(var)2-10 in PGCs gave rise to a tumorous phenotype of germline cells in ovaries. Because this phenotype indicates loss of female identity of germline cells, we consider Su(var)2-10 to be a strong candidate target of Sxl in PGCs. Our results represent a first step toward elucidating the Sxl-dependent feminizing pathway in the germline.
Collapse
Affiliation(s)
- Ryoma Ota
- Life Science Center of Tsukuba Advanced Research Alliance (TARA Center), University of Tsukuba, Tsukuba, 305-8577, Japan
| | - Shumpei Morita
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, 305-8572, Japan
| | - Masanao Sato
- Laboratory of Applied Molecular Entomology, Division of Applied Bioscience, Graduate School of Agriculture, Hokkaido University, Sapporo, 060-8589, Japan
| | - Shuji Shigenobu
- Functional Genomics Facility, NIBB Core Research Facilities, National Institute for Basic Biology, Nishigo-naka 38, Myodaiji, Okazaki, 444-8585, Japan
| | - Makoto Hayashi
- Life Science Center of Tsukuba Advanced Research Alliance (TARA Center), University of Tsukuba, Tsukuba, 305-8577, Japan
| | - Satoru Kobayashi
- Life Science Center of Tsukuba Advanced Research Alliance (TARA Center), University of Tsukuba, Tsukuba, 305-8577, Japan.,Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, 305-8572, Japan
| |
Collapse
|
28
|
Bombyx mori P-element Somatic Inhibitor (BmPSI) Is a Key Auxiliary Factor for Silkworm Male Sex Determination. PLoS Genet 2017; 13:e1006576. [PMID: 28103247 PMCID: PMC5289617 DOI: 10.1371/journal.pgen.1006576] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 02/02/2017] [Accepted: 01/10/2017] [Indexed: 12/23/2022] Open
Abstract
Manipulation of sex determination pathways in insects provides the basis for a wide spectrum of strategies to benefit agriculture and public health. Furthermore, insects display a remarkable diversity in the genetic pathways that lead to sex differentiation. The silkworm, Bombyx mori, has been cultivated by humans as a beneficial insect for over two millennia, and more recently as a model system for studying lepidopteran genetics and development. Previous studies have identified the B. mori Fem piRNA as the primary female determining factor and BmMasc as its downstream target, while the genetic scenario for male sex determination was still unclear. In the current study, we exploite the transgenic CRISPR/Cas9 system to generate a comprehensive set of knockout mutations in genes BmSxl, Bmtra2, BmImp, BmImpM, BmPSI and BmMasc, to investigate their roles in silkworm sex determination. Absence of Bmtra2 results in the complete depletion of Bmdsx transcripts, which is the conserved downstream factor in the sex determination pathway, and induces embryonic lethality. Loss of BmImp or BmImpM function does not affect the sexual differentiation. Mutations in BmPSI and BmMasc genes affect the splicing of Bmdsx and the female reproductive apparatus appeared in the male external genital. Intriguingly, we identify that BmPSI regulates expression of BmMasc, BmImpM and Bmdsx, supporting the conclusion that it acts as a key auxiliary factor in silkworm male sex determination. The sex determination system extremely diverse among organisms including insects in which even each order occupy a different manner of sex determination. The silkworm, Bombyx mori, is a lepidopteran model insect with economic importance. The mechanism of the silkworm sex determination has been in mystery for a long time until a Fem piRNA was identified as the primary female sex determinator recently. However, genetic and phenotypic proofs are urgently needed to fully exploit the mechanism, especially of the male sex determination. In the current study, we provided comprehensively genetic evidences by generating CRISPR/Cas9-mediated knockout mutations for those genes BmSxl, Bmtra2, BmImp, BmImpM, BmPSI and BmMasc, which were considered to be involved in insect sex determination. The results showed that mutations of BmSxl, BmImp and BmImpM had no physiological and morphological effects on the sexual development while Bmtra2 depletion caused Bmdsx splicing disappeared and induced embryonic lethality. Importantly, the BmPSI regulates expression of BmMasc, BmImpM and Bmdsx, supporting the conclusion that it acts as a key auxiliary factor to regulate the male sex determination in the silkworm.
Collapse
|
29
|
Zhang XL, Wu J, Wang J, Shen T, Li H, Lu J, Gu Y, Kang Y, Wong CH, Ngan CY, Shao Z, Wu J, Zhao X. Integrative epigenomic analysis reveals unique epigenetic signatures involved in unipotency of mouse female germline stem cells. Genome Biol 2016; 17:162. [PMID: 27465593 PMCID: PMC4963954 DOI: 10.1186/s13059-016-1023-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/08/2016] [Indexed: 01/26/2023] Open
Abstract
Background Germline stem cells play an essential role in establishing the fertility of an organism. Although extensively characterized, the regulatory mechanisms that govern the fundamental properties of mammalian female germline stem cells remain poorly understood. Results We generate genome-wide profiles of the histone modifications H3K4me1, H3K27ac, H3K4me3, and H3K27me3, DNA methylation, and RNA polymerase II occupancy and perform transcriptome analysis in mouse female germline stem cells. Comparison of enhancer regions between embryonic stem cells and female germline stem cells identifies the lineage-specific enhancers involved in germline stem cell features. Additionally, our results indicate that DNA methylation primarily contributes to female germline stem cell unipotency by suppressing the somatic program and is potentially involved in maintenance of sexual identity when compared with male germline stem cells. Moreover, we demonstrate down-regulation of Prmt5 triggers differentiation and thus uncover a role for Prmt5 in maintaining the undifferentiated status of female germline stem cells. Conclusions The genome-wide epigenetic signatures and the transcription regulators identified here provide an invaluable resource for understanding the fundamental features of mouse female germline stem cells. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-1023-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiao-Li Zhang
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jun Wu
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jian Wang
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Tingting Shen
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hua Li
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jun Lu
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yunzhao Gu
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yani Kang
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chee-Hong Wong
- Sequencing Technology Group, Joint Genome Institute, Lawrence Berkeley National Laboratory, Walnut Creek, CA, 94598, USA
| | - Chew Yee Ngan
- Sequencing Technology Group, Joint Genome Institute, Lawrence Berkeley National Laboratory, Walnut Creek, CA, 94598, USA
| | - Zhifeng Shao
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ji Wu
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China. .,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| | - Xiaodong Zhao
- Shanghai Center for Systems Biomedicine, Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
30
|
Wang JQ, Cao WG. Key Signaling Events for Committing Mouse Pluripotent Stem Cells to the Germline Fate. Biol Reprod 2015; 94:24. [PMID: 26674564 DOI: 10.1095/biolreprod.115.135095] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/07/2015] [Indexed: 01/01/2023] Open
Abstract
The process of germline development carries genetic information and preparatory totipotency across generations. The last decade has witnessed remarkable successes in the generation of germline cells from mouse pluripotent stem cells, especially induced germline cells with the capacity for producing viable offspring, suggesting clinical applications of induced germline cells in humans. However, to date, the culture systems for germline induction with accurate sex-specific meiosis and epigenetic reprogramming have not been well-established. In this study, we primarily focus on the mouse model to discuss key signaling events for germline induction. We review mechanisms of competent regulators on primordial germ cell induction and discuss current achievements and difficulties in inducing sex-specific germline development. Furthermore, we review the developmental identities of mouse embryonic stem cells and epiblast stem cells under certain defined culture conditions as it relates to the differentiation process of becoming germline cells.
Collapse
Affiliation(s)
- Jian-Qi Wang
- Transgenic and Stem Cell Core, Institute of Animal Sciences and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wen-Guang Cao
- Transgenic and Stem Cell Core, Institute of Animal Sciences and Veterinary Medicine, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
31
|
Liu W, Li SZ, Li Z, Wang Y, Li XY, Zhong JX, Zhang XJ, Zhang J, Zhou L, Gui JF. Complete depletion of primordial germ cells in an All-female fish leads to Sex-biased gene expression alteration and sterile All-male occurrence. BMC Genomics 2015; 16:971. [PMID: 26582363 PMCID: PMC4652418 DOI: 10.1186/s12864-015-2130-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/22/2015] [Indexed: 01/05/2023] Open
Abstract
Background Gynogenesis is one of unisexual reproduction modes in vertebrates, and produces all-female individuals with identical genetic background. In sexual reproduction vertebrates, the roles of primordial germ cells on sexual dimorphism and gonadal differentiation have been largely studied, and two distinct functional models have been proposed. However, the role of primordial germ cells remains unknown in unisexual animals, and it is also unclear whether the functional models in sexual reproduction animals are common in unisexual animals. Results To solve these puzzles, we attempt to utilize the gynogenetic superiority of polyploid Carassius gibelio to create a complete germ cell-depleted gonad model by a similar morpholino-mediated knockdown approach used in other examined sexual reproduction fishes. Through the germ cell-depleted gonad model, we have performed comprehensive and comparative transcriptome analysis, and revealed a complete alteration of sex-biased gene expression. Moreover, the expression alteration leads to up-regulation of testis-biased genes and down-regulation of ovary-biased genes, and results in the occurrence of sterile all-males with testis-like gonads and secondary sex characteristics in the germ cell-depleted gynogenetic Carassius gibelio. Conclusions Our current results have demonstrated that unisexual gynogenetic embryos remain keeping male sex determination information in the genome, and the complete depletion of primordial germ cells in the all-female fish leads to sex-biased gene expression alteration and sterile all-male occurrence. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2130-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Shi-Zhu Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Zhi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Xi-Yin Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jian-Xiang Zhong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Xiao-Juan Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jun Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China
| | - Jian-Fang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Graduate University of the Chinese Academy of Sciences, Wuhan, 430072, China.
| |
Collapse
|
32
|
Tower J. Mitochondrial maintenance failure in aging and role of sexual dimorphism. Arch Biochem Biophys 2015; 576:17-31. [PMID: 25447815 PMCID: PMC4409928 DOI: 10.1016/j.abb.2014.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/08/2014] [Accepted: 10/18/2014] [Indexed: 12/31/2022]
Abstract
Gene expression changes during aging are partly conserved across species, and suggest that oxidative stress, inflammation and proteotoxicity result from mitochondrial malfunction and abnormal mitochondrial-nuclear signaling. Mitochondrial maintenance failure may result from trade-offs between mitochondrial turnover versus growth and reproduction, sexual antagonistic pleiotropy and genetic conflicts resulting from uni-parental mitochondrial transmission, as well as mitochondrial and nuclear mutations and loss of epigenetic regulation. Aging phenotypes and interventions are often sex-specific, indicating that both male and female sexual differentiation promote mitochondrial failure and aging. Studies in mammals and invertebrates implicate autophagy, apoptosis, AKT, PARP, p53 and FOXO in mediating sex-specific differences in stress resistance and aging. The data support a model where the genes Sxl in Drosophila, sdc-2 in Caenorhabditis elegans, and Xist in mammals regulate mitochondrial maintenance across generations and in aging. Several interventions that increase life span cause a mitochondrial unfolded protein response (UPRmt), and UPRmt is also observed during normal aging, indicating hormesis. The UPRmt may increase life span by stimulating mitochondrial turnover through autophagy, and/or by inhibiting the production of hormones and toxic metabolites. The data suggest that metazoan life span interventions may act through a common hormesis mechanism involving liver UPRmt, mitochondrial maintenance and sexual differentiation.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Program, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089-2910, United States.
| |
Collapse
|
33
|
König A, Shcherbata HR. Soma influences GSC progeny differentiation via the cell adhesion-mediated steroid-let-7-Wingless signaling cascade that regulates chromatin dynamics. Biol Open 2015; 4:285-300. [PMID: 25661868 PMCID: PMC4359735 DOI: 10.1242/bio.201410553] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It is known that signaling from the germline stem cell niche is required to maintain germline stem cell identity in Drosophila. However, it is not clear whether the germline stem-cell daughters differentiate by default (because they are physically distant from the niche) or whether additional signaling is necessary to initiate the differentiation program. Previously, we showed that ecdysteroid signaling cell non-autonomously regulates early germline differentiation via its soma-specific co-activator and co-repressor, Taiman and Abrupt. Now, we demonstrate that this regulation is modulated by the miRNA let-7, which acts in a positive feedback loop to confer ecdysone signaling robustness via targeting its repressor, the transcription factor Abrupt. This feedback loop adjusts ecdysteroid signaling in response to some stressful alterations in the external and internal conditions, which include temperature stress and aging, but not nutritional deprivation. Upon let-7 deficit, escort cells fail to properly differentiate: their shape, division, and cell adhesive characteristics are perturbed. These cells have confused cellular identity and form columnar-like rather than squamous epithelium and fail to send protrusions in between differentiating germline cysts, affecting soma-germline communication. Particularly, levels of the homophilic cell adhesion protein Cadherin, which recruits Wg signaling transducer β-catenin, are increased in mutant escort cells and, correspondingly, in the adjacent germline cells. Readjustment of heterotypic (soma-germline) cell adhesion modulates Wg signaling intensity in the germline, which in turn regulates histone modifications that promote expression of the genes necessary to trigger early germline differentiation. Thus, our data first show the intrinsic role for Wg signaling in the germline and support a model where the soma influences the tempo of germline differentiation in response to external conditions.
Collapse
Affiliation(s)
- Annekatrin König
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Halyna R Shcherbata
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany
| |
Collapse
|
34
|
Bachtrog D, Mank JE, Peichel CL, Kirkpatrick M, Otto SP, Ashman TL, Hahn MW, Kitano J, Mayrose I, Ming R, Perrin N, Ross L, Valenzuela N, Vamosi JC. Sex determination: why so many ways of doing it? PLoS Biol 2014; 12:e1001899. [PMID: 24983465 PMCID: PMC4077654 DOI: 10.1371/journal.pbio.1001899] [Citation(s) in RCA: 744] [Impact Index Per Article: 67.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Sexual reproduction is an ancient feature of life on earth, and the familiar X and Y chromosomes in humans and other model species have led to the impression that sex determination mechanisms are old and conserved. In fact, males and females are determined by diverse mechanisms that evolve rapidly in many taxa. Yet this diversity in primary sex-determining signals is coupled with conserved molecular pathways that trigger male or female development. Conflicting selection on different parts of the genome and on the two sexes may drive many of these transitions, but few systems with rapid turnover of sex determination mechanisms have been rigorously studied. Here we survey our current understanding of how and why sex determination evolves in animals and plants and identify important gaps in our knowledge that present exciting research opportunities to characterize the evolutionary forces and molecular pathways underlying the evolution of sex determination.
Collapse
Affiliation(s)
- Doris Bachtrog
- University of California, Berkeley, Department of Integrative Biology, Berkeley, California, United States of America
| | - Judith E. Mank
- University College London, Department of Genetics, Evolution and Environment, London, United Kingdom
| | - Catherine L. Peichel
- Fred Hutchinson Cancer Research Center, Divisions of Human Biology and Basic Sciences, Seattle, Washington, United States of America
| | - Mark Kirkpatrick
- University of Texas, Department of Integrative Biology, Austin, Texas, United States of America
| | - Sarah P. Otto
- University of British Columbia, Department of Zoology, Vancouver, British Columbia, Canada
| | - Tia-Lynn Ashman
- University of Pittsburgh, Department of Biological Sciences, Pittsburgh, Pennsylvania, United States of America
| | - Matthew W. Hahn
- Indiana University, Department of Biology, Bloomington Indiana, United States of America
| | - Jun Kitano
- National Institute of Genetics, Ecological Genetics Laboratory, Mishima, Shizuoka, Japan
| | - Itay Mayrose
- Tel Aviv University, Department of Molecular Biology and Ecology of Plants, Tel Aviv, Israel
| | - Ray Ming
- University of Illinois, Department of Plant Biology, Urbana-Champaign, Illinois, United States of America
| | - Nicolas Perrin
- University of Lausanne, Department of Ecology and Evolution, Lausanne, Switzerland
| | - Laura Ross
- University of Oxford, Department of Zoology, Oxford, United Kingdom
| | - Nicole Valenzuela
- Iowa State University, Department of Ecology, Evolution and Organismal Biology, Ames, Iowa, United States of America
| | - Jana C. Vamosi
- University of Calgary, Department of Biological Sciences, Calgary, Alberta, Canada
| | | |
Collapse
|
35
|
Functional analysis of the Drosophila embryonic germ cell transcriptome by RNA interference. PLoS One 2014; 9:e98579. [PMID: 24896584 PMCID: PMC4045815 DOI: 10.1371/journal.pone.0098579] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 05/05/2014] [Indexed: 11/19/2022] Open
Abstract
In Drosophila melanogaster, primordial germ cells are specified at the posterior pole of the very early embryo. This process is regulated by the posterior localized germ plasm that contains a large number of RNAs of maternal origin. Transcription in the primordial germ cells is actively down-regulated until germ cell fate is established. Bulk expression of the zygotic genes commences concomitantly with the degradation of the maternal transcripts. Thus, during embryogenesis, maternally provided and zygotically transcribed mRNAs determine germ cell development collectively. In an effort to identify novel genes involved in the regulation of germ cell behavior, we carried out a large-scale RNAi screen targeting both maternal and zygotic components of the embryonic germ line transcriptome. We identified 48 genes necessary for distinct stages in germ cell development. We found pebble and fascetto to be essential for germ cell migration and germ cell division, respectively. Our data uncover a previously unanticipated role of mei-P26 in maintenance of embryonic germ cell fate. We also performed systematic co-RNAi experiments, through which we found a low rate of functional redundancy among homologous gene pairs. As our data indicate a high degree of evolutionary conservation in genetic regulation of germ cell development, they are likely to provide valuable insights into the biology of the germ line in general.
Collapse
|
36
|
Rosner A, Moiseeva E, Rabinowitz C, Rinkevich B. Germ lineage properties in the urochordate Botryllus schlosseri - from markers to temporal niches. Dev Biol 2013; 384:356-74. [PMID: 24120376 DOI: 10.1016/j.ydbio.2013.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 08/25/2013] [Accepted: 10/03/2013] [Indexed: 01/28/2023]
Abstract
The primordial germ cells (PGCs) in the colonial urochordate Botryllus schlosseri are sequestered in late embryonic stage. PGC-like populations, located at any blastogenic stage in specific niches, inside modules with curtailed lifespan, survive throughout the life of the colony by repeated weekly migration to newly formed buds. This cyclical migration and the lack of specific markers for PGC-like populations are obstacles to the study on PGCs. For that purpose, we isolated the Botryllus DDX1 (BS-DDX1) and characterized it by normal expression patterns and by specific siRNA knockdown experiments. Expression of BS-DDX1 concurrent with BS-Vasa, γ-H2AX, BS-cadherin and phospho-Smad1/5/8, demarcate PGC cells from soma cells and from more differentiated germ cells lineages, which enabled the detection of additional putative transient niches in zooids. Employing BS-cadherin siRNA knockdown, retinoic acid (RA) administration or β-estradiol administration affirmed the BS-Vasa(+)BS-DDX1(+)BS-cadherin(+)γ-H2AX(+)phospho-Smad1/5/8(+) population as the B. schlosseri PGC-like cells. By striving to understand the PGC-like cells trafficking between transient niches along blastogenic cycles, CM-DiI-stained PGC-like enriched populations from late blastogenic stage D zooids were injected into genetically matched colonial ramets at blastogenic stages A or C and their fates were observed for 9 days. Based on the accumulated data, we conceived a novel network of several transient and short lived 'germ line niches' that preserve PGCs homeostasis, protecting these cells from the weekly astogenic senescence processes, thus enabling the survival of the PGCs throughout the organism's life.
Collapse
Affiliation(s)
- Amalia Rosner
- National Institute of Oceanography, Israel Oceanography & Limnological Research, Tel Shikmona, P.O. Box 8030, Haifa 31080, Israel.
| | | | | | | |
Collapse
|
37
|
Abstract
Drosophilists have identified many, or perhaps most, of the key regulatory genes determining sex using classical genetics, however, regulatory genes must ultimately result in the deployment of the genome in a quantitative manner, replete with complex interactions with other regulatory pathways. In the last decade, genomics has provided a rich picture of the transcriptional profile of the sexes that underlies sexual dimorphism. The current challenge is linking transcriptional profiles with the regulatory genes. This will be a complex synthesis, but the prospects for progress are outstanding.
Collapse
Affiliation(s)
- Emily Clough
- Section of Developmental Genomics and Laboratory of Cellular and Developmental Biology, National Institute of Diabetes, and Digestive and Kidney Diseases, National Institutes of Health, Bethesda MD 20892-8028, USA.
| | | |
Collapse
|
38
|
Abstract
The Drosophila Sex-lethal (Sxl) gene encodes a female-specific RNA binding protein that in somatic cells globally regulates all aspects of female-specific development and behavior. Sxl also has a critical, but less well understood, role in female germ cells. Germ cells without Sxl protein can adopt a stem cell fate when housed in a normal ovary, but fail to successfully execute the self-renewal differentiation fate switch. The failure to differentiate is accompanied by the inappropriate expression of a set of male specific markers, continued proliferation, and formation of a tumor. The findings in Chau et al., (2012) identify the germline stem cell maintenance factor nanos as one of its target genes, and suggest that Sxl enables the switch from germline stem cell to committed daughter cell by posttranscriptional downregulation of nanos expression. These studies provide the basis for a new model in which Sxl directly couples sexual identity with the self-renewal differentiation decision and raises several interesting questions about the genesis of the tumor phenotype.
Collapse
Affiliation(s)
- Helen K Salz
- Department of Genetics and Genome Sciences, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
39
|
SACY-1 DEAD-Box helicase links the somatic control of oocyte meiotic maturation to the sperm-to-oocyte switch and gamete maintenance in Caenorhabditis elegans. Genetics 2012; 192:905-28. [PMID: 22887816 PMCID: PMC3522166 DOI: 10.1534/genetics.112.143271] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In sexually reproducing animals, oocytes arrest at diplotene or diakinesis and resume meiosis (meiotic maturation) in response to hormones. In Caenorhabditis elegans, major sperm protein triggers meiotic resumption through a mechanism involving somatic Gαs–adenylate cyclase signaling and soma-to-germline gap-junctional communication. Using genetic mosaic analysis, we show that the major effector of Gαs–adenylate cyclase signaling, protein kinase A (PKA), is required in gonadal sheath cells for oocyte meiotic maturation and dispensable in the germ line. This result rules out a model in which cyclic nucleotides must transit through sheath-oocyte gap junctions to activate PKA in the germ line, as proposed in vertebrate systems. We conducted a genetic screen to identify regulators of oocyte meiotic maturation functioning downstream of Gαs–adenylate cyclase–PKA signaling. We molecularly identified 10 regulatory loci, which include essential and nonessential factors. sacy-1, which encodes a highly conserved DEAD-box helicase, is an essential germline factor that negatively regulates meiotic maturation. SACY-1 is a multifunctional protein that establishes a mechanistic link connecting the somatic control of meiotic maturation to germline sex determination and gamete maintenance. Modulatory factors include multiple subunits of a CoREST-like complex and the TWK-1 two-pore potassium channel. These factors are not absolutely required for meiotic maturation or its negative regulation in the absence of sperm, but function cumulatively to enable somatic control of meiotic maturation. This work provides insights into the genetic control of meiotic maturation signaling in C. elegans, and the conserved factors identified here might inform analysis in other systems through either homology or analogy.
Collapse
|
40
|
Yang SY, Baxter EM, Van Doren M. Phf7 controls male sex determination in the Drosophila germline. Dev Cell 2012; 22:1041-51. [PMID: 22595675 DOI: 10.1016/j.devcel.2012.04.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 03/05/2012] [Accepted: 04/13/2012] [Indexed: 01/11/2023]
Abstract
Establishment of germline sexual identity is critical for production of male and female germline stem cells, as well as sperm versus eggs. Here we identify PHD Finger Protein 7 (PHF7) as an important factor for male germline sexual identity in Drosophila. PHF7 exhibits male-specific expression in early germ cells, germline stem cells, and spermatogonia. It is important for germline stem cell maintenance and gametogenesis in males, whereas ectopic expression in female germ cells ablates the germline. Strikingly, expression of PHF7 promotes spermatogenesis in XX germ cells when they are present in a male soma. PHF7 homologs are also specifically expressed in the mammalian testis, and human PHF7 rescues Drosophila Phf7 mutants. PHF7 associates with chromatin, and both the human and fly proteins bind histone H3 N-terminal tails with a preference for dimethyl lysine 4 (H3K4me2). We propose that PHF7 acts as a conserved epigenetic "reader" that activates the male germline sexual program.
Collapse
Affiliation(s)
- Shu Yuan Yang
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | |
Collapse
|
41
|
Kopp A. Dmrt genes in the development and evolution of sexual dimorphism. Trends Genet 2012; 28:175-84. [PMID: 22425532 DOI: 10.1016/j.tig.2012.02.002] [Citation(s) in RCA: 200] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 02/07/2012] [Accepted: 02/08/2012] [Indexed: 12/20/2022]
Abstract
Most animals are sexually dimorphic, but different taxa have different sex-specific traits. Despite major differences in the genetic control of sexual development among animal lineages, the doublesex/mab-3 related (Dmrt) family of transcription factors has been shown to be involved in sex-specific differentiation in all animals that have been studied. In recent years the functions of Dmrt genes have been characterized in many animal groups, opening the way to a broad comparative perspective. This review focuses on the similarities and differences in the functions of Dmrt genes across the animal kingdom. I highlight a number of common themes in the sexual development of different taxa, discuss how Dmrt genes have acquired new roles during animal evolution, and show how they have contributed to the origin of novel sex-specific traits.
Collapse
Affiliation(s)
- Artyom Kopp
- Department of Evolution and Ecology, University of California-Davis, Davis, CA 95616 USA.
| |
Collapse
|
42
|
Abstract
Cell-cell signaling and adhesion are critical for establishing tissue architecture during development and for maintaining tissue architecture and function in the adult. Defects in adhesion and signaling can result in mislocalization of cells, uncontrolled proliferation and improper differentiation, leading to tissue overgrowth, tumor formation, and cancer metastasis. An important example is found in the germline. Germ cells that are not incorporated into the gonad exhibit a greater propensity for forming germ cell tumors, and defects in germline development can reduce fertility. While much attention is given to germ cells, their development into functional gametes depends upon somatic gonadal cells. The study of model organisms has provided great insights into how somatic gonadal cells are specified, the molecular mechanisms that regulate gonad morphogenesis, and the role of germline-soma communication in the establishment and maintenance of the germline stem cell niche. This work will be discussed in the context of Drosophila melanogaster.
Collapse
Affiliation(s)
- Jennifer C Jemc
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
43
|
Losick VP, Morris LX, Fox DT, Spradling A. Drosophila stem cell niches: a decade of discovery suggests a unified view of stem cell regulation. Dev Cell 2011; 21:159-71. [PMID: 21763616 PMCID: PMC6894370 DOI: 10.1016/j.devcel.2011.06.018] [Citation(s) in RCA: 241] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 06/03/2011] [Accepted: 06/10/2011] [Indexed: 12/28/2022]
Abstract
The past decade of research on Drosophila stem cells and niches has provided key insights. Fly stem cells do not occupy a special "state" based on novel "stem cell genes" but resemble transiently arrested tissue progenitors. Moreover, individual stem cells and downstream progenitors are highly dynamic and dispensable, not tissue bulwarks. Niches, rather than fixed cell lineages, ensure tissue health by holding stem cells and repressing cell differentiation inside, but not outside. We review the five best-understood adult Drosophila stem cells and argue that the fundamental biology of stem cells and niches is conserved between Drosophila and mice.
Collapse
Affiliation(s)
- Vicki P Losick
- Howard Hughes Medical Institute Research Laboratories, Department of Embryology, Carnegie Institution for Science, 3520 San Martin Drive, Baltimore, MD 21218, USA
| | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Mark Van Doren
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
45
|
Hashiyama K, Hayashi Y, Kobayashi S. Drosophila Sex lethal gene initiates female development in germline progenitors. Science 2011; 333:885-8. [PMID: 21737698 DOI: 10.1126/science.1208146] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Sex determination in the Drosophila germ line is regulated by both the sex of the surrounding soma and cell-autonomous cues. How primordial germ cells (PGCs) initiate sexual development via cell-autonomous mechanisms is unclear. Here, we demonstrate that, in Drosophila, the Sex lethal (Sxl) gene acts autonomously in PGCs to induce female development. Sxl is transiently expressed in PGCs during their migration to the gonads; this expression, which was detected only in XX PGCs, is necessary for PGCs to assume a female fate. Ectopic expression of Sxl in XY PGCs was sufficient to induce them to enter oogenesis and produce functional eggs when transplanted into an XX host. Our data provide powerful evidence that Sxl initiates female germline fate during sexual development.
Collapse
Affiliation(s)
- Kazuya Hashiyama
- Okazaki Institute for Integrative Bioscience, National Institute for Basic Biology, National Institutes of Natural Sciences, Higashiyama, Myodaiji, Okazaki 444-8787, Japan
| | | | | |
Collapse
|