1
|
Sreejith P, Kim C. Lin28 is Required for Single Niche Development in the Drosophila Male Gonad. Dev Reprod 2023; 27:221-226. [PMID: 38292237 PMCID: PMC10824566 DOI: 10.12717/dr.2023.27.4.221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/22/2023] [Accepted: 11/23/2023] [Indexed: 02/01/2024]
Abstract
A stem cell niche provides an environment that governs stem cell maintenance and division. Thus, the development of a proper niche is of prime importance to stem cell behaviors. Mechanisms of niche development are beginning to be revealed in the Drosophila male gonad. Niche cells are initially dispersed throughout the gonad, then assemble at its apical tip through the anterior migration of posteriorly located niche cells. The molecular mechanisms of this migration and assembly are still poorly understood. Here we show evidence suggesting that Lin28, an RNA-binding protein and regulator of let7 genesis, might be an intrinsic factor for the anterior migration of niche cells. We found that a dispersed, ectopic niche, a phenotype observed with anterior migration defects, occurs in lin28 mutant gonads. This phenotype is rescued by expression of lin28 in the niche cells. These findings suggest that Lin28 might be required for the anterior migration of niche cells.
Collapse
Affiliation(s)
- Perinthottathil Sreejith
- Department of Biomedical Genetics,
University of Rochester Medical Center, Rochester,
NY 14642, USA
| | - Changsoo Kim
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| |
Collapse
|
2
|
Zhao J, Sun X, Dai H, Zhang X, Zhang D, Zhu X. Changes in Gene Expression of Whiteflies, Bemisia tabaci MED Feeding on Tomato Plants Infected by One of the Criniviruses, Tomato Chlorosis Virus through Transcriptome Analysis. Int J Genomics 2023; 2023:3807812. [PMID: 37261104 PMCID: PMC10228217 DOI: 10.1155/2023/3807812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/12/2022] [Accepted: 05/01/2023] [Indexed: 06/02/2023] Open
Abstract
Tomato chlorosis virus (ToCV), transmitted by the whitefly, Bemisia tabaci (Gennadius; Hemiptera: Aleyrodidae) has been continuously emerging on tomato plants and causing a significant economic loss throughout China. In the current study, RNA-Seq analysis was used to explore the gene expression profiles of B. tabaci Mediterranean (MED) that fed on both ToCV-infected and -uninfected tomato plants for 6, 12, 24, and 48 hours, respectively. The results revealed that dynamic changes occurred in the gene expressions of whiteflies at different time intervals after they acquired the virus. A total of 1709, 461, 4548, and 1748 differentially expressed genes (DEGs) were identified after a 6, 12, 24, and 48 hours feeding interval for the viral acquisition, respectively. The least number of expressed genes appeared in whiteflies with the 12 hours feeding treatment, and the largest numbers of those found in those with 24 hours feeding treatment. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that B. tabaci MED responded to ToCV acquisition through altering its nerve system development, fertility, detoxification, glucose metabolism, and immune function before it lost its ability to transmit the virus. The number of DEGs, degree of differential gene expressions, expression level of the same gene, involved biological processes, and metabolic functions in whiteflies post the 12 hours feeding, and viral acquisition were different from those from other three feeding treatments, which could be a significant finding suggesting an effective control of B. tabaci MED should be done less than 12 hours after whiteflies started feeding on ToCV-infected tomatoes. Our results further provided a clarified understanding in how B. tabaci was protected from viral acquisitions through comparison of the differential profile of gene expressions in whiteflies feeding on plants that were infected by semipersistent viruses.
Collapse
Affiliation(s)
- Jing Zhao
- Key Laboratory of Biology and Molecular Biology of University in Shandong, College of Seed and Facility Agricultural Engineering, Weifang University, Weifang 261061, China
| | - Xiaoan Sun
- Facility Horticulture of University in Shandong, College of Agriculture, Weifang University of Science & Technology, Shouguang 262700, China
| | - Huijie Dai
- Facility Horticulture of University in Shandong, College of Agriculture, Weifang University of Science & Technology, Shouguang 262700, China
| | - Xianping Zhang
- Shandong Provincial key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| | - Dezhen Zhang
- Facility Horticulture of University in Shandong, College of Agriculture, Weifang University of Science & Technology, Shouguang 262700, China
| | - Xiaoping Zhu
- Shandong Provincial key Laboratory for Biology of Vegetable Diseases and Insect Pests, College of Plant Protection, Shandong Agricultural University, Taian 271018, China
| |
Collapse
|
3
|
Raz AA, Vida GS, Stern SR, Mahadevaraju S, Fingerhut JM, Viveiros JM, Pal S, Grey JR, Grace MR, Berry CW, Li H, Janssens J, Saelens W, Shao Z, Hu C, Yamashita YM, Przytycka T, Oliver B, Brill JA, Krause H, Matunis EL, White-Cooper H, DiNardo S, Fuller MT. Emergent dynamics of adult stem cell lineages from single nucleus and single cell RNA-Seq of Drosophila testes. eLife 2023; 12:e82201. [PMID: 36795469 PMCID: PMC9934865 DOI: 10.7554/elife.82201] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/19/2023] [Indexed: 02/17/2023] Open
Abstract
Proper differentiation of sperm from germline stem cells, essential for production of the next generation, requires dramatic changes in gene expression that drive remodeling of almost all cellular components, from chromatin to organelles to cell shape itself. Here, we provide a single nucleus and single cell RNA-seq resource covering all of spermatogenesis in Drosophila starting from in-depth analysis of adult testis single nucleus RNA-seq (snRNA-seq) data from the Fly Cell Atlas (FCA) study. With over 44,000 nuclei and 6000 cells analyzed, the data provide identification of rare cell types, mapping of intermediate steps in differentiation, and the potential to identify new factors impacting fertility or controlling differentiation of germline and supporting somatic cells. We justify assignment of key germline and somatic cell types using combinations of known markers, in situ hybridization, and analysis of extant protein traps. Comparison of single cell and single nucleus datasets proved particularly revealing of dynamic developmental transitions in germline differentiation. To complement the web-based portals for data analysis hosted by the FCA, we provide datasets compatible with commonly used software such as Seurat and Monocle. The foundation provided here will enable communities studying spermatogenesis to interrogate the datasets to identify candidate genes to test for function in vivo.
Collapse
Affiliation(s)
- Amelie A Raz
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Howard Hughes Medical InstituteCambridgeUnited States
| | - Gabriela S Vida
- Department of Cell and Developmental Biology, The Perelman School of Medicine and The Penn Institute for Regenerative MedicinePhiladelphiaUnited States
| | - Sarah R Stern
- Department of Developmental Biology, Stanford University School of MedicineStanfordUnited States
| | - Sharvani Mahadevaraju
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaUnited States
| | - Jaclyn M Fingerhut
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Howard Hughes Medical InstituteCambridgeUnited States
| | - Jennifer M Viveiros
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Soumitra Pal
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of HealthBethesdaUnited States
| | - Jasmine R Grey
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Mara R Grace
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Cameron W Berry
- Department of Developmental Biology, Stanford University School of MedicineStanfordUnited States
| | - Hongjie Li
- Huffington Center on Aging and Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Jasper Janssens
- JVIB Center for Brain & Disease Research, and the Department of Human Genetics, KU LeuvenLeuvenBelgium
| | - Wouter Saelens
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, and Department of Applied Mathematics, Computer Science and Statistics, Ghent UniversityGhentBelgium
| | - Zhantao Shao
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Chun Hu
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Yukiko M Yamashita
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Howard Hughes Medical InstituteCambridgeUnited States
| | - Teresa Przytycka
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of HealthBethesdaUnited States
| | - Brian Oliver
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaUnited States
| | - Julie A Brill
- Cell Biology Program, The Hospital for Sick ChildrenTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
- Institute of Medical Science, University of TorontoTorontoCanada
| | - Henry Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Erika L Matunis
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | | | - Stephen DiNardo
- Department of Cell and Developmental Biology, The Perelman School of Medicine and The Penn Institute for Regenerative MedicinePhiladelphiaUnited States
| | - Margaret T Fuller
- Department of Developmental Biology, Stanford University School of MedicineStanfordUnited States
- Department of Genetics, Stanford UniversityStanfordUnited States
| |
Collapse
|
4
|
Anllo L, DiNardo S. Visceral mesoderm signaling regulates assembly position and function of the Drosophila testis niche. Dev Cell 2022; 57:1009-1023.e5. [PMID: 35390292 PMCID: PMC9050945 DOI: 10.1016/j.devcel.2022.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022]
Abstract
Tissue homeostasis often requires a properly placed niche to support stem cells. Morphogenetic processes that position a niche are just being described. For the Drosophila testis, we recently showed that pro-niche cells, specified at disparate positions during early gonadogenesis, must assemble into one collective at the anterior of the gonad. We now find that Slit and FGF signals emanating from adjacent visceral mesoderm regulate assembly. In response to signaling, niche cells express islet, which we find is also required for niche assembly. Without signaling, niche cells specified furthest from the anterior are unable to migrate, remaining dispersed. The function of such niches is severely disrupted, with niche cells evading cell cycle quiescence, compromised in their ability to signal the incipient stem cell pool, and failing to orient stem cell divisions properly. Our work identifies both extrinsic signaling and intrinsic responses required for proper assembly and placement of the testis niche.
Collapse
Affiliation(s)
- Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| |
Collapse
|
5
|
Shore T, Levi T, Kalifa R, Dreifuss A, Rekler D, Weinberg-Shukron A, Nevo Y, Bialistoky T, Moyal V, Gold MY, Leebhoff S, Zangen D, Deshpande G, Gerlitz O. Nucleoporin107 mediates female sexual differentiation via Dsx. eLife 2022; 11:72632. [PMID: 35311642 PMCID: PMC8975549 DOI: 10.7554/elife.72632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 03/17/2022] [Indexed: 11/13/2022] Open
Abstract
We recently identified a missense mutation in Nucleoporin107 (Nup107; D447N) underlying XX-ovarian-dysgenesis, a rare disorder characterized by underdeveloped and dysfunctional ovaries. Modeling of the human mutation in Drosophila or specific knockdown of Nup107 in the gonadal soma resulted in ovarian-dysgenesis-like phenotypes. Transcriptomic analysis identified the somatic sex-determination gene doublesex (dsx) as a target of Nup107. Establishing Dsx as a primary relevant target of Nup107, either loss or gain of Dsx in the gonadal soma is sufficient to mimic or rescue the phenotypes induced by Nup107 loss. Importantly, the aberrant phenotypes induced by compromising either Nup107 or dsx are reminiscent of BMP signaling hyperactivation. Remarkably, in this context, the metalloprotease AdamTS-A, a transcriptional target of both Dsx and Nup107, is necessary for the calibration of BMP signaling. As modulation of BMP signaling is a conserved critical determinant of soma-germline interaction, the sex and tissue specific deployment of Dsx-F by Nup107 seems crucial for the maintenance of the homeostatic balance between the germ cells and somatic gonadal cells.
Collapse
Affiliation(s)
- Tikva Shore
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Tgst Levi
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Rachel Kalifa
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Amatzia Dreifuss
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Dina Rekler
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | | | - Yuval Nevo
- Bioinformatics Unit of the I-CORE Computation Center, The Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Tzofia Bialistoky
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Victoria Moyal
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Merav Yaffa Gold
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - Shira Leebhoff
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| | - David Zangen
- Division of Pediatric Endocrinology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Girish Deshpande
- Department of Molecular Biology, Princeton University, Princeton, United States
| | - Offer Gerlitz
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem, Israel
| |
Collapse
|
6
|
Grmai L, Pozmanter C, Van Doren M. The Regulation of Germline Sex Determination in Drosophila by Sex lethal. Sex Dev 2022; 16:323-328. [PMID: 35259743 PMCID: PMC10540089 DOI: 10.1159/000521235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/29/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The establishment of male or female identity (sex determination) is essential for creating the anatomical, physiological, and behavioral differences between 2 sexes of the same species (sexual dimorphism). In many organisms, including mammals and Drosophila, sex is determined by inheritance of sex chromosomes, while in other animals, sex is determined by environmental factors. Arguably the most important consequence of sex determination is the production of healthy gametes necessary for reproduction: female oocytes and male spermatids. SUMMARY The generation of sperm and oocytes requires cooperation between 2 different cell types within the gonad: germ cells and somatic cells. Defects in sex determination in either the somatic gonad or germline lead to disorders of sexual development and infertility. In Drosophila, the gene Sex lethal (Sxl) is the key determinant of sex in both the soma and the germline. However, how Sxl controls sex determination is much more well understood in the soma than the germline. Key Mesage: This review will focus on Sxl in the germline, how it is activated specifically in female germ cells, and how it regulates germline sex determination and sexual development.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Caitlin Pozmanter
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mark Van Doren
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
7
|
Matsushima D, Kasahara R, Matsuno K, Aoki F, Suzuki MG. Involvement of Ecdysone Signaling in the Expression of the doublesex Gene during Embryonic Development in the Silkworm, Bombyx mori. Sex Dev 2019; 13:151-163. [PMID: 31487710 DOI: 10.1159/000502361] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2019] [Indexed: 01/09/2023] Open
Abstract
Steroid hormones, represented by estrogen and testosterone, act as sex hormones that play an essential role in the sexual differentiation of vertebrates. However, it remains unclear whether ecdysteroids, typical steroid hormones in insects, function as sex hormones. In this study, we investigated whether ecdysteroids or ecdysone signals are involved in the sexual differentiation of the silkworm (Bombyx mori) embryo. Quantitative analysis using LC-MS/MS demonstrated that there was no significant difference in the 20-hydroxyecdysone (20E) titer between sexes during embryonic development. Consistent with this result, expression levels of 2 genes encoding ecdysteroid-phosphate phosphatase (EPPase) and ecdysone 20-hydroxylase (E20OHase), which are essential for the biosynthesis of ecdysone and 20E in eggs, did not show a significant difference between male and female embryos. Expression levels of ecdysone receptor (EcR) and E75, which is one of a small set of genes induced directly by 20E, were also similar between the 2 sexes. However, knockdown of EPPase and one isoform of EcR (EcR-A) resulted in decreased expression of Bombyx doublesex (Bmdsx), a master regulatory gene for sexual differentiation of the silkworm in both male and female embryos. In vitro analysis with cultured testes revealed that expression levels of Bmdsx were increased in a dose-dependent manner of the ecdysone analog, ponasterone A. These results suggest that ecdysone signaling may play a role in indirectly regulating the expression of some genes involved in sexual differentiation through inducing expression of Bmdsx in the silkworm.
Collapse
|
8
|
Camara N, Whitworth C, Dove A, Van Doren M. Doublesex controls specification and maintenance of the gonad stem cell niches in Drosophila. Development 2019; 146:dev.170001. [PMID: 31043421 DOI: 10.1242/dev.170001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 04/23/2019] [Indexed: 11/20/2022]
Abstract
Sex-specific development of the gonads is a key aspect of sexual dimorphism that is regulated by Doublesex/Mab3-related transcription factors (DMRTs) in diverse animal species. We find that in mutants for Drosophila dsx, important components of the male and female gonad stem cell niches (hubs and terminal filaments/cap cells, respectively) still form. Initially, gonads in all dsx mutants (both XX and XY) initiate the male program of development, but later half of these gonads switch to form female stem cell niche structures. One individual can have both male-type and female-type gonad niches; however, male and female niches are usually not observed in the same gonad, indicating that cells make a 'group decision' about which program to follow. We conclude that dsx does not act in an instructive manner to regulate male versus female niche formation, as these structures form in the absence of dsx function. Instead, dsx acts to 'tip the balance' between the male or female programs, which are then executed independently of dsx We show that bric a brac acts downstream of dsx to control the male versus female niche decision. These results indicate that, in both flies and mammals, the sexual fate of the somatic gonad is remarkably plastic and is controlled by a combination of autonomous and non-autonomous cues.
Collapse
Affiliation(s)
- Nicole Camara
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Cale Whitworth
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Abigail Dove
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Mark Van Doren
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
9
|
Li X, Jin B, Dong Y, Chen X, Tu Z, Gu J. Two of the three Transformer-2 genes are required for ovarian development in Aedes albopictus. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2019; 109:92-105. [PMID: 30914323 PMCID: PMC6636634 DOI: 10.1016/j.ibmb.2019.03.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/18/2019] [Accepted: 03/21/2019] [Indexed: 05/23/2023]
Abstract
In Drosophila melanogaster, transformer-2 (tra2) plays an essential role in the sex-specific splicing of doublesex (dsx) and fruitless (fru), two key transcription factor genes that program sexual differentiation and regulate sexual behavior. In the present study, the sequences and expression profiles of three tra2 (Aalbtra2) genes in the Asian tiger mosquito, Aedes albopictus (Ae. albopictus) were characterized. Phylogenetic analysis revealed that these paralogs resulted from two duplication events. The first occurred in the common ancestor of Culicidae, giving rise to the tra2-α and tra2-β clades that are found across divergent mosquito genera, including Aedes, Culex, and Anopheles. The second occurred within the tra2-α clade, giving rise to tra2-γ in Ae. albopictus. In addition to the conserved RNA recognition motif (RRM), arginine-rich/serine-rich regions (RS domains) and a linker region, a glycine-rich region located between the RRM and RS2 was observed in Tra2-α and Tra2-γ of Ae. albopictus that has not yet been described in the Tra2 proteins of dipteran insects. Quantitative real-time PCR detected relatively high levels of transcripts from all three tra2 paralogs in 0-2 h embryos, suggesting maternal deposition of these transcripts. All three Aalbtra2 genes were highly expressed in the ovary, while Aalbtra2-β was also highly expressed in the testis. RNAi-mediated knockdown of any or all Aalbtra2 genes did not result in an obvious switch of the sex-specificity in dsx and fru splicing in the whole-body samples. However, knockdown of transcripts from all three tra2 genes significantly reduced the female isoform of dsx mRNA and increased the male isoform of the dsx mRNA in both the ovary and the fat body in adult females. Furthermore, knockdown of either Aalbtra2-α or Aalbtra2-γ or all three Aalbtra2 led to a decrease in ovariole number and ovary size after a blood meal. Taken together, these results indicate that two of the three tra2 genes affect female ovarian development.
Collapse
Affiliation(s)
- Xiaocong Li
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Binbin Jin
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yunqiao Dong
- Reproductive Medical Centre of Guangdong Women and Children Hospital, Guangzhou, Guangdong, 511442, China
| | - Xiaoguang Chen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Zhijian Tu
- Department of Biochemistry, Virginia Tech, Blacksburg, VA, 24060, USA
| | - Jinbao Gu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
10
|
Genome-Wide Identification and Expression Profiling of Wnt Family Genes in the Silkworm, Bombyx mori. Int J Mol Sci 2019; 20:ijms20051221. [PMID: 30862048 PMCID: PMC6429082 DOI: 10.3390/ijms20051221] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 03/04/2019] [Accepted: 03/06/2019] [Indexed: 02/07/2023] Open
Abstract
Wnt is a family of conserved glycoproteins that participate in a variety of important biological processes including embryo development, cell proliferation and differentiation, and tissue regeneration. The Wnt family is a metazoan novelty found in all animal phyla. Studies have revealed that the number of Wnt genes varies among species, presumably due to reproduction and loss of genes during evolution. However, a comprehensive inventory of Wnt genes in Lepidoptera is lacking. In this study, we identified the repertoire of Wnt genes in the silkworm and seven other species of Lepidoptera and obtained eight Wnt genes (Wnt1, Wnt5–Wnt7, Wnt9–Wnt11, and WntA) in each species. Four of these Wnt genes are clustered in two orientations (5′-Wnt9-Wnt1-Wnt6-Wnt10-3′ and 5′-Wnt10-Wnt6-Wnt1-Wnt9-3′) in both moths and butterflies. Transcript analysis of Wnt in silkworm embryonic stages showed that each BmWnt gene had a unique expression pattern during embryological development. Analysis of a larval stage revealed differential expression of Wnt family members in diverse tissues. Our study provides an overview of the Wnt family in Lepidoptera and will inspire further functional study of the Wnt genes in the silkworm.
Collapse
|
11
|
Anllo L, Plasschaert LW, Sui J, DiNardo S. Live imaging reveals hub cell assembly and compaction dynamics during morphogenesis of the Drosophila testis niche. Dev Biol 2018; 446:102-118. [PMID: 30553808 DOI: 10.1016/j.ydbio.2018.12.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/19/2018] [Accepted: 12/12/2018] [Indexed: 01/23/2023]
Abstract
Adult stem cells are often found in specialized niches, where the constituent cells direct self-renewal of their stem cell pool. The niche is therefore crucial for both normal homeostasis and tissue regeneration. In many mammalian tissues, niche cells have classically been difficult to identify, which has hampered any understanding of how tissues first construct niches during development. Fortunately, the Drosophila germline stem cell (GSC) niche is well defined, allowing for unambiguous identification of both niche cells and resident stem cells. The testis niche first forms in the early embryo, during a late stage of gonadogenesis. Here, using live-imaging both in vivo and ex vivo, we follow pro-niche cells as they assemble and assume their final form. We show that after ex vivo culture the niche appears fully functional, as judged by enrichment of adhesion proteins, the ability to activate STAT in adjacent GSCs, and to direct GSCs to divide orthogonally to the niche, just as they would in situ. Collectively, our imaging has generated several novel insights on niche morphogenesis that could not be inferred from fixed images alone. We identify dynamic processes that constitute an assembly phase and a compaction phase during morphogenesis. The compaction phase correlates with cell neighbor exchange among the assembled pro-niche cells, as well as a burst of divisions among newly recruited stem cells. Before compaction, an assembly phase involves the movement of pro-niche cells along the outer periphery of the gonad, using the extracellular matrix (ECM) to assemble at the anterior of the gonad. Finally, live-imaging in integrin mutants allows us to define the role of pro-niche cell-ECM interaction with regard to the new assembly and compaction dynamics revealed here.
Collapse
Affiliation(s)
- Lauren Anllo
- The Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| | - Lindsey W Plasschaert
- The Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| | - Justin Sui
- The Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| | - Stephen DiNardo
- The Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| |
Collapse
|
12
|
Effect of dietary energy restriction and subsequent compensatory feeding on testicular transcriptome in developing rams. Theriogenology 2018; 119:198-207. [DOI: 10.1016/j.theriogenology.2018.06.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 06/28/2018] [Accepted: 06/28/2018] [Indexed: 11/15/2022]
|
13
|
Saari S, Andjelković A, Garcia GS, Jacobs HT, Oliveira MT. Expression of Ciona intestinalis AOX causes male reproductive defects in Drosophila melanogaster. BMC DEVELOPMENTAL BIOLOGY 2017; 17:9. [PMID: 28673232 PMCID: PMC5496232 DOI: 10.1186/s12861-017-0151-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 06/14/2017] [Indexed: 02/08/2023]
Abstract
Background Mitochondrial alternative respiratory-chain enzymes are phylogenetically widespread, and buffer stresses affecting oxidative phosphorylation in species that possess them. However, they have been lost in the evolutionary lineages leading to vertebrates and arthropods, raising the question as to what survival or reproductive disadvantages they confer. Recent interest in using them in therapy lends a biomedical dimension to this question. Methods Here, we examined the impact of the expression of Ciona intestinalis alternative oxidase, AOX, on the reproductive success of Drosophila melanogaster males. Sperm-competition assays were performed between flies carrying three copies of a ubiquitously expressed AOX construct, driven by the α-tubulin promoter, and wild-type males of the same genetic background. Results In sperm-competition assays, AOX conferred a substantial disadvantage, associated with decreased production of mature sperm. Sperm differentiation appeared to proceed until the last stages, but was spatially deranged, with spermatozoids retained in the testis instead of being released to the seminal vesicle. High AOX expression was detected in the outermost cell-layer of the testis sheath, which we hypothesize may disrupt a signal required for sperm maturation. Conclusions AOX expression in Drosophila thus has effects that are deleterious to male reproductive function. Our results imply that AOX therapy must be developed with caution. Electronic supplementary material The online version of this article (doi:10.1186/s12861-017-0151-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sina Saari
- Institute of Biosciences and Medical Technology, Tampere University Hospital, University of Tampere, FI-33014, Tampere, Finland
| | - Ana Andjelković
- Institute of Biosciences and Medical Technology, Tampere University Hospital, University of Tampere, FI-33014, Tampere, Finland
| | - Geovana S Garcia
- Departamento de Tecnologia, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista "Júlio de Mesquita Filho", Jaboticabal, SP, 14884-900, Brazil
| | - Howard T Jacobs
- Institute of Biosciences and Medical Technology, Tampere University Hospital, University of Tampere, FI-33014, Tampere, Finland. .,Institute of Biotechnology, University of Helsinki, FI-00014, Helsinki, Finland.
| | - Marcos T Oliveira
- Institute of Biosciences and Medical Technology, Tampere University Hospital, University of Tampere, FI-33014, Tampere, Finland.,Departamento de Tecnologia, Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista "Júlio de Mesquita Filho", Jaboticabal, SP, 14884-900, Brazil
| |
Collapse
|
14
|
Estrogen related receptor is required for the testicular development and for the normal sperm axoneme/mitochondrial derivatives in Drosophila males. Sci Rep 2017; 7:40372. [PMID: 28094344 PMCID: PMC5240334 DOI: 10.1038/srep40372] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 12/06/2016] [Indexed: 12/22/2022] Open
Abstract
Estrogen related receptors (ERRs), categorized as orphan nuclear receptors, are critical for energy homeostasis and somatic development. However, significance of ERRs in the development of reproductive organs/organelles/cells remain poorly understood, albeit their homology to estrogen receptors. In this context, here, we show that knockdown of ERR in the testes leads to improperly developed testes with mis-regulation of genes (aly, mia, bruce, bam, bgcn, fzo and eya) involved in spermatogenesis, resulting in reduced male fertility. The observed testicular deformity is consistent with the down-regulation of SOX-E group of gene (SOX100B) in Drosophila. We also show dispersion/disintegration of fusomes (microtubule based structures associated with endoplasmic reticulum derived vesicle, interconnecting spermatocytes) in ERR knockdown testes. A few ERR knockdown testes go through spermatogenesis but have significantly fewer sperm. Moreover, flagella of these sperm are defective with abnormal axoneme and severely reduced mitochondrial derivatives, suggesting a possible role for ERR in mitochondrial biogenesis, analogous to mammalian ERRα. Interestingly, similar knockdown of remaining seventeen nuclear receptors did not yield a detectable reproductive or developmental defect in Drosophila. These findings add newer dimensions to the functions envisaged for ERR and provide the foundation for deciphering the relevance of orphan nuclear receptors in ciliopathies and testicular dysgenesis.
Collapse
|
15
|
Prakash A, Monteiro A. Molecular mechanisms of secondary sexual trait development in insects. CURRENT OPINION IN INSECT SCIENCE 2016; 17:40-48. [PMID: 27720072 DOI: 10.1016/j.cois.2016.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/04/2016] [Accepted: 06/06/2016] [Indexed: 06/06/2023]
Abstract
Secondary sexual traits are those traits other than the primary gametes that distinguish the sexes of a species. The development of secondary sexual traits occurs when sexually dimorphic factors, that is, molecules differentially produced by primary sex determination systems in males and females, are integrated into the gene regulatory networks responsible for sexual trait development. In insects, these molecular asymmetric factors were always considered to originate inside the trait-building cells, but recent work points to external factors, such as hormones, as potential candidates mediating secondary sexual trait development. Here, we review examples of the different molecular mechanisms producing sexually dimorphic traits in insects, and suggest a need to revise our understanding of secondary sexual trait development within the insect lineage.
Collapse
Affiliation(s)
- Anupama Prakash
- Department of Biological Sciences, National University of Singapore, Singapore.
| | - Antónia Monteiro
- Department of Biological Sciences, National University of Singapore, Singapore; Yale-NUS College, Singapore.
| |
Collapse
|
16
|
Monahan AJ, Starz-Gaiano M. Apontic regulates somatic stem cell numbers in Drosophila testes. BMC DEVELOPMENTAL BIOLOGY 2016; 16:5. [PMID: 26993259 PMCID: PMC4799534 DOI: 10.1186/s12861-016-0103-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 02/10/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Microenvironments called niches maintain resident stem cell populations by balancing self-renewal with differentiation, but the genetic regulation of this process is unclear. The niche of the Drosophila testis is well-characterized and genetically tractable, making it ideal for investigating the molecular regulation of stem cell biology. The JAK/STAT pathway, activated by signals from a niche component called the hub, maintains both germline and somatic stem cells. RESULTS This study investigated the molecular regulation of the JAK/STAT pathway in the stem cells of the Drosophila testis. We determined that the transcriptional regulator Apontic (Apt) acts in the somatic (cyst) stem cells (CySCs) to balance differentiation and maintenance. We found Apt functions as a negative feedback inhibitor of STAT activity, which enables cyst cell maturation. Simultaneous loss of the STAT regulators apt and Socs36E, or the Stat92E-targeting microRNA miR-279, expanded the somatic stem cell-like population. CONCLUSIONS Genetic analysis revealed that a conserved genetic regulatory network limits JAK/STAT activity in the somatic stem cells of Drosophila testis. In these cells, we determined JAK/STAT signaling promotes apt expression. Then, Apt functions through Socs36E and miR-279 to attenuate pathway activation, which is required for timely CySC differentiation. We propose that Apt acts as a core component of a STAT-regulatory circuit to prevent stem cell overpopulation and allow stem cell maturation.
Collapse
Affiliation(s)
- Amanda J Monahan
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA.,Present Address: Department of Medicine, Division of Infectious Disease, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Michelle Starz-Gaiano
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA.
| |
Collapse
|
17
|
Rideout EJ, Narsaiya MS, Grewal SS. The Sex Determination Gene transformer Regulates Male-Female Differences in Drosophila Body Size. PLoS Genet 2015; 11:e1005683. [PMID: 26710087 PMCID: PMC4692505 DOI: 10.1371/journal.pgen.1005683] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 10/28/2015] [Indexed: 12/17/2022] Open
Abstract
Almost all animals show sex differences in body size. For example, in Drosophila, females are larger than males. Although Drosophila is widely used as a model to study growth, the mechanisms underlying this male-female difference in size remain unclear. Here, we describe a novel role for the sex determination gene transformer (tra) in promoting female body growth. Normally, Tra is expressed only in females. We find that loss of Tra in female larvae decreases body size, while ectopic Tra expression in males increases body size. Although we find that Tra exerts autonomous effects on cell size, we also discovered that Tra expression in the fat body augments female body size in a non cell-autonomous manner. These effects of Tra do not require its only known targets doublesex and fruitless. Instead, Tra expression in the female fat body promotes growth by stimulating the secretion of insulin-like peptides from insulin producing cells in the brain. Our data suggest a model of sex-specific growth in which body size is regulated by a previously unrecognized branch of the sex determination pathway, and identify Tra as a novel link between sex and the conserved insulin signaling pathway. Female-biased sexual size dimorphism is common in invertebrates, yet the mechanisms underlying increased female body size remain unclear. We uncovered a key role for sex determination gene transformer (tra) in promoting increased growth in females. Interestingly, we found that sex differences in body size are regulated by Tra in a pathway that is separate of the canonical sex determination pathway, and of other aspects of sexual dimorphism. Instead, Tra function in the fat body regulates growth in a non cell-autonomous manner by regulating the secretion of insulin-like peptides from the brain. This novel Tra-insulin link we describe may have implications for other sexually dimorphic phenotypes in Drosophila (eg. lifespan, stress resistance), many of which are also regulated by insulin.
Collapse
Affiliation(s)
- Elizabeth J. Rideout
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail: (EJR); (SSG)
| | - Marcus S. Narsaiya
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Savraj S. Grewal
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, Alberta, Canada
- * E-mail: (EJR); (SSG)
| |
Collapse
|
18
|
Abstract
The embryonic gonad of Drosophila melanogaster begins to display sexually dimorphic traits soon after its formation. Here we demonstrate the involvement of a wnt family ligand, wnt-2, in the induction of these sex-specific differences. We show that wnt-2 contributes to the survival of a male-specific population of somatic gonadal precursor cells (SGPs), the male-specific SGPs that are located at the posterior of the male gonad. We also show that the Wnt-2 ligand synergizes with the JAK-STAT ligand Upd, which is produced by SGPs at the anterior of the gonad to activate the STAT pathway in male germ cells. We suggest that the use of two spatially separated signaling systems to initiate the JAK-STAT stem cell maintenance pathway in germ cells provides a mechanism for increasing the pool of potential progenitors of the germline stem cells in the adult testes. Finally, we present evidence indicating that, like the JAK-STAT pathway, wnt-2 stimulates germ cells in male embryos to re-enter the cell cycle.
Collapse
|
19
|
Schardt L, Ander JJ, Lohmann I, Papagiannouli F. Stage-specific control of niche positioning and integrity in the Drosophila testis. Mech Dev 2015; 138 Pt 3:336-48. [PMID: 26226434 DOI: 10.1016/j.mod.2015.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/23/2015] [Accepted: 07/24/2015] [Indexed: 12/11/2022]
Abstract
A fundamental question is how complex structures are maintained after their initial specification. Stem cells reside in a specialized microenvironment, called niche, which provides essential signals controlling stem cell behavior. We addressed this question by studying the Drosophila male stem cell niche, called the hub. Once specified, the hub cells need to maintain their position and architectural integrity through embryonic, larval and pupal stages of testis organogenesis and during adult life. The Hox gene Abd-B, in addition to its described role in male embryonic gonads, maintains the architecture and positioning of the larval hub from the germline by affecting integrin localization in the neighboring somatic cyst cells. We find that the AbdB-Boss/Sev cascade affects integrin independent of Talin, while genetic interactions depict integrin as the central downstream player in this system. Focal adhesion and integrin-adaptor proteins within the somatic stem cells and cyst cells, such as Paxillin, Pinch and Vav, also contribute to proper hub integrity and positioning. During adult stages, hub positioning is controlled by Abd-B activity in the outer acto-myosin sheath, while Abd-B expression in adult spermatocytes exerts no effect on hub positioning and integrin localization. Our data point at a cell- and stage-specific function of Abd-B and suggest that the occurrence of new cell types and cell interactions in the course of testis organogenesis made it necessary to adapt the whole system by reusing the same players for male stem cell niche positioning and integrity in an alternative manner.
Collapse
Affiliation(s)
- Lisa Schardt
- Centre for Organismal Studies (COS) Heidelberg, Cell Networks - Cluster of Excellence, University of Heidelberg, D-69120, Germany; Deutsches Krebsforschungszentrum (DKFZ), D-69120, Germany
| | - Janina-Jacqueline Ander
- Centre for Organismal Studies (COS) Heidelberg, Cell Networks - Cluster of Excellence, University of Heidelberg, D-69120, Germany
| | - Ingrid Lohmann
- Centre for Organismal Studies (COS) Heidelberg, Cell Networks - Cluster of Excellence, University of Heidelberg, D-69120, Germany.
| | - Fani Papagiannouli
- Centre for Organismal Studies (COS) Heidelberg, Cell Networks - Cluster of Excellence, University of Heidelberg, D-69120, Germany.
| |
Collapse
|
20
|
Morphological allometry and intersexuality in horsehair-worm-infected mantids, Hierodula formosana (Mantodea: Mantidae). Parasitology 2015; 142:1130-42. [PMID: 25891833 DOI: 10.1017/s0031182015000360] [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] [Indexed: 11/08/2022]
Abstract
Parasitic castration is a strategy used by parasites to minimize damage to the host by consuming its reproductive system, which results in the morphological alteration of the host. We determined that the forewing shape and density of the antennal sensilla of field-collected adult male mantids (Hierodula formosana), infected by horsehair worms (Chordodes formosanus) was partially feminized (intersexuality), and both male and female mantids infected by horsehair worms exhibited allometric changes in their wings and walking legs. In addition, the testes of most infected male adults disappeared or reduced in size, whereas the number of ovarioles in infected female adults was unaffected. The infection mainly influenced the structures related to host reproduction and locomotion, suggesting unbalanced energy exploitation and the reduction of parasitic virulence. In addition, the intersexuality of infected male adults indicated that sexual differentiation in insects, which researchers have considered to be an autonomous process, was influenced by the infection. The similarity of the antennae of infected male adults with those of last-instar female nymphs suggested that parasitic juvenilization may cause such feminization, but the mechanism of parasitic influence on insect sex characteristics should be studied further.
Collapse
|
21
|
Sex- and tissue-specific functions of Drosophila doublesex transcription factor target genes. Dev Cell 2015; 31:761-73. [PMID: 25535918 DOI: 10.1016/j.devcel.2014.11.021] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 10/02/2014] [Accepted: 11/13/2014] [Indexed: 11/20/2022]
Abstract
Primary sex-determination "switches" evolve rapidly, but Doublesex (DSX)-related transcription factors (DMRTs) act downstream of these switches to control sexual development in most animal species. Drosophila dsx encodes female- and male-specific isoforms (DSX(F) and DSX(M)), but little is known about how dsx controls sexual development, whether DSX(F) and DSX(M) bind different targets, or how DSX proteins direct different outcomes in diverse tissues. We undertook genome-wide analyses to identify DSX targets using in vivo occupancy, binding site prediction, and evolutionary conservation. We find that DSX(F) and DSX(M) bind thousands of the same targets in multiple tissues in both sexes, yet these targets have sex- and tissue-specific functions. Interestingly, DSX targets show considerable overlap with targets identified for mouse DMRT1. DSX targets include transcription factors and signaling pathway components providing for direct and indirect regulation of sex-biased expression.
Collapse
|
22
|
Papagiannouli F, Lohmann I. Stage-specific control of stem cell niche architecture in the Drosophila testis by the posterior Hox gene Abd-B. Comput Struct Biotechnol J 2015; 13:122-30. [PMID: 25750700 PMCID: PMC4348433 DOI: 10.1016/j.csbj.2015.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/12/2015] [Accepted: 01/15/2015] [Indexed: 11/30/2022] Open
Abstract
A fundamental question in biology is how complex structures are maintained after their initial specification. We address this question by reviewing the role of the Hox gene Abd-B in Drosophila testis organogenesis, which proceeds through embryonic, larval and pupal stages to reach maturation in adult stages. The data presented in this review highlight a cell- and stage-specific function of Abd-B, since the mechanisms regulating stem cell niche positioning and architecture at different stages seem to be different despite the employment of similar factors. In addition to its described role in the male embryonic gonads, sustained activity of Abd-B in the pre-meiotic germline spermatocytes during larval stages is required to maintain the architecture of the stem cell niche by regulating βPS-integrin localization in the neighboring somatic cyst cells. Loss of Abd-B is associated with cell non-autonomous effects within the niche, leading to a dramatic reduction of pre-meiotic cell populations in adult testes. Identification of Abd-B target genes revealed that Abd-B mediates its effects by controlling the activity of the sevenless ligand Boss via its direct targets Src42A and Sec63. During adult stages, when testis morphogenesis is completed with the addition of the acto-myosin sheath originating from the genital disc, stem cell niche positioning and integrity are regulated by Abd-B activity in the acto-myosin sheath whereas integrin acts in an Abd-B independent way. It seems that the occurrence of new cell types and cell interactions in the course of testis organogenesis made it necessary to adapt the system to the new cellular conditions by reusing the same players for testis stem cell niche positioning in an alternative manner.
Collapse
Affiliation(s)
- Fani Papagiannouli
- Centre for Organismal Studies (COS) Heidelberg, Cell Networks - Cluster of Excellence, University of Heidelberg, D-69120, Germany
| | - Ingrid Lohmann
- Centre for Organismal Studies (COS) Heidelberg, Cell Networks - Cluster of Excellence, University of Heidelberg, D-69120, Germany
| |
Collapse
|
23
|
Hamm RL, Meisel RP, Scott JG. The evolving puzzle of autosomal versus Y-linked male determination in Musca domestica. G3 (BETHESDA, MD.) 2014; 5:371-84. [PMID: 25552607 PMCID: PMC4349091 DOI: 10.1534/g3.114.014795] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 12/27/2014] [Indexed: 11/18/2022]
Abstract
Sex determination is one of the most rapidly evolving developmental pathways, but the factors responsible for this fast evolution are not well resolved. The house fly, Musca domestica, is an ideal model for studying sex determination because house fly sex determination is polygenic and varies considerably between populations. Male house flies possess a male-determining locus, the M factor, which can be located on the Y or X chromosome or any of the five autosomes. There can be a single M or multiple M factors present in an individual male, in heterozygous or homozygous condition. Males with multiple copies of M skew the sex ratio toward the production of males. Potentially in response to these male-biased sex ratios, an allele of the gene transformer, Md-tra(D), promotes female development in the presence of one or multiple M factors. There have been many studies to determine the linkage and frequency of these male determining factors and the frequency of Md-tra(D) chromosomes in populations from around the world. This review provides a summary of the information available to date regarding the patterns of distribution of autosomal, X-linked and Y-linked M factors, the relative frequencies of the linkage of M, the changes in frequencies found in field populations, and the fitness of males with autosomal M factors vs. Y-linked M. We evaluate this natural variation in the house fly sex determination pathway in light of models of the evolution of sex determination.
Collapse
Affiliation(s)
| | - Richard P Meisel
- Department of Biology and Biochemistry, University of Houston, Houston, Texas 77204
| | - Jeffrey G Scott
- Department of Entomology, Comstock Hall, Cornell University, Ithaca, New York 14853
| |
Collapse
|
24
|
Whitworth C, Jimenez E, Van Doren M. Development of sexual dimorphism in the Drosophila testis. SPERMATOGENESIS 2014; 2:129-136. [PMID: 23087832 PMCID: PMC3469436 DOI: 10.4161/spmg.21780] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The creation of sexual dimorphism in the gonads is essential for producing the male and female gametes required for sexual reproduction. Sexual development of the gonads involves both somatic cells and germ cells, which often undergo sex determination by different mechanisms. While many sex-specific characteristics evolve rapidly and are very different between animal species, gonad function and the formation of sperm and eggs appear more similar and may be more conserved. Consistent with this, the doublesex/mab3 Related Transcription factors (DMRTs) are important for gonad sexual dimorphism in a wide range of animals, including flies, worms and mammals. Here we explore how sexual dimorphism is regulated in the Drosophila gonad, focusing on recent discoveries relating to testis development. We will discuss how sex determination in both the germline and the soma are utilized to create a testis, including the role of the key somatic sex determination factor doublesex.
Collapse
Affiliation(s)
- Cale Whitworth
- Department of Biology; The Johns Hopkins University; Baltimore, MD USA
| | | | | |
Collapse
|
25
|
Fidler A, Boulay L, Wawersik M. Sonication-facilitated immunofluorescence staining of late-stage embryonic and larval Drosophila tissues in situ. J Vis Exp 2014:e51528. [PMID: 25146311 PMCID: PMC4827934 DOI: 10.3791/51528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Studies performed in Drosophila melanogaster embryos and larvae provide crucial insight into developmental processes such as cell fate specification and organogenesis. Immunostaining allows for the visualization of developing tissues and organs. However, a protective cuticle that forms at the end of embryogenesis prevents permeation of antibodies into late-stage embryos and larvae. While dissection prior to immunostaining is regularly used to analyze Drosophila larval tissues, it proves inefficient for some analyses because small tissues may be difficult to locate and isolate. Sonication provides an alternative to dissection in larval Drosophila immunostaining protocols. It allows for quick, simultaneous processing of large numbers of late-stage embryos and larvae and maintains in situ morphology. After fixation in formaldehyde, a sample is sonicated. Sample is then subjected to immunostaining with antigen-specific primary antibodies and fluorescently labeled secondary antibodies to visualize target cell types and specific proteins via fluorescence microscopy. During the process of sonication, proper placement of a sonicating probe above the sample, as well as the duration and intensity of sonication, is critical. Additonal minor modifications to standard immunostaining protocols may be required for high quality stains. For antibodies with low signal to noise ratio, longer incubation times are typically necessary. As a proof of concept for this sonication-facilitated protocol, we show immunostains of three tissue types (testes, ovaries, and neural tissues) at a range of developmental stages.
Collapse
|
26
|
Santerre C, Sourdaine P, Adeline B, Martinez AS. Cg-SoxE and Cg-β-catenin, two new potential actors of the sex-determining pathway in a hermaphrodite lophotrochozoan, the Pacific oyster Crassostrea gigas. Comp Biochem Physiol A Mol Integr Physiol 2014; 167:68-76. [DOI: 10.1016/j.cbpa.2013.09.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 09/27/2013] [Accepted: 09/30/2013] [Indexed: 10/26/2022]
|
27
|
Sinden D, Badgett M, Fry J, Jones T, Palmen R, Sheng X, Simmons A, Matunis E, Wawersik M. Jak-STAT regulation of cyst stem cell development in the Drosophila testis. Dev Biol 2012; 372:5-16. [PMID: 23010510 DOI: 10.1016/j.ydbio.2012.09.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 09/15/2012] [Accepted: 09/16/2012] [Indexed: 10/27/2022]
Abstract
Establishment and maintenance of functional stem cells is critical for organ development and tissue homeostasis. Little is known about the mechanisms underlying stem establishment during organogenesis. Drosophila testes are among the most thoroughly characterized systems for studying stem cell behavior, with germline stem cells (GSCs) and somatic cyst stem cells (CySCs) cohabiting a discrete stem cell niche at the testis apex. GSCs and CySCs are arrayed around hub cells that also comprise the niche and communication between hub cells, GSCs, and CySCs regulates the balance between stem cell maintenance and differentiation. Recent data has shown that functional, asymmetrically dividing GSCs are first established at ∼23 h after egg laying during Drosophila testis morphogenesis (Sheng et al., 2009). This process correlates with coalescence of the hub, but development of CySCs from somatic gonadal precursors (SGPs) was not examined. Here, we show that functional CySCs are present at the time of GSC establishment, and that Jak-STAT signaling is necessary and sufficient for CySC maintenance shortly thereafter. Furthermore, hyper-activation of Jak in CySCs promotes expansion of the GSC population, while ectopic Jak activation in the germline induces GSC gene expression in GSC daughter cells but does not prevent spermatogenic differentiation. Together, these observations indicate that, similar to adult testes, Jak-STAT signaling from the hub acts on both GSCs and CySC to regulate their development and differentiation, and that additional signaling from CySCs to the GSCs play a dominant role in controlling GSC maintenance during niche formation.
Collapse
Affiliation(s)
- D Sinden
- College of William & Mary, Biology Department, Williamsburg, VA 23185, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Renault AD. vasa is expressed in somatic cells of the embryonic gonad in a sex-specific manner in Drosophila melanogaster. Biol Open 2012; 1:1043-8. [PMID: 23213382 PMCID: PMC3507172 DOI: 10.1242/bio.20121909] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 07/31/2012] [Indexed: 11/28/2022] Open
Abstract
Vasa is a DEAD box helicase expressed in the Drosophila germline at all stages of development. vasa homologs are found widely in animals and vasa has become the gene of choice in identifying germ cells. I now show that Drosophila vasa expression is not restricted to the germline but is also expressed in a somatic lineage, the embryonic somatic gonadal precursor cells. This expression is sexually dimorphic, being maintained specifically in males, and is regulated post-transcriptionally. Although somatic Vasa expression is not required for gonad coalescence, these data support the notion that Vasa is not solely a germline factor.
Collapse
Affiliation(s)
- Andrew D Renault
- Max Planck Institute for Developmental Biology , Spemannstrasse 35, 72076 Tübingen , Germany
| |
Collapse
|
29
|
Papagiannouli F, Lohmann I. Shaping the niche: lessons from the Drosophila testis and other model systems. Biotechnol J 2012; 7:723-36. [PMID: 22488937 DOI: 10.1002/biot.201100352] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 01/31/2012] [Accepted: 02/27/2012] [Indexed: 11/12/2022]
Abstract
Stem cells are fascinating, as they supply the cells that construct our adult bodies and replenish, as we age, worn out, damaged, and diseased tissues. Stem cell regulation relies on intrinsic signals but also on inputs emanating from the neighbouring niche. The Drosophila testis provides an excellent system for studying such processes. Although recent advances have uncovered several signalling, cytoskeletal and other factors affecting niche homeostasis and testis differentiation, many aspects of niche regulation and maintenance remain unsolved. In this review, we discuss aspects of niche establishment and integrity not yet fully understood and we compare it to the current knowledge in other model systems such as vertebrates and plants. We also address specific questions on stem cell maintenance and niche regulation in the Drosophila testis under the control of Hox genes. Finally, we provide insights on the striking functional conservation of homologous genes in plants and animals and their respective stem cell niches. Elucidating conserved mechanisms of stem cell control in both lineages could reveal the importance underlying this conservation and justify the evolutionary pressure to adapt homologous molecules for performing the same task.
Collapse
Affiliation(s)
- Fani Papagiannouli
- Centre for Organismal Studies (COS) Heidelberg and CellNetworks - Cluster of Excellence, Heidelberg, Germany.
| | | |
Collapse
|
30
|
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: 210] [Impact Index Per Article: 16.2] [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
|
31
|
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
|
32
|
Negri I, Pellecchia M, Grève P, Daffonchio D, Bandi C, Alma A. Sex and stripping: The key to the intimate relationship between Wolbachia and host? Commun Integr Biol 2011; 3:110-5. [PMID: 20585501 DOI: 10.4161/cib.3.2.10520] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 11/03/2009] [Indexed: 11/19/2022] Open
Abstract
Wolbachia pipientis is known to infect only arthropods and nematodes (mainly filarial worms). A unique feature shared by the two Phyla is the ability to replace the exoskeleton, a process known as ecdysis. This shared characteristic is thought to reflect a common ancestry. Arthropod moulting is induced by the steroid hormone 20-hydroxyecdysone (20E) and a role for ecdysteroids in nematode ecdysis has also been suggested. Removing Wolbachia from filarial worms impairs the host's development. From analyses of the genome of Wolbachia harbored by the filarial nematode Brugia malayi and that of its host, the bacterium may provide a source of heme, an essential component of cytochrome P450's that are necessary for steroid hormone biosynthetic pathways.In arthropods, Wolbachia is a reproductive manipulator, inducing various phenotypic effects that may be due to differences in host physiology, in particular, endocrine-related processes governing development and reproduction. Insect steroids have well-defined roles in the coordination of multiple developmental processes, and in adults they control important aspects of reproduction, including ovarian development, oogenesis, sexual behavior, and in some taxa vitellogenin biosynthesis.According to some authors ecdysteroids may also act as sex hormones. In insects sex differentiation is generally thought to be a strictly genetic process, in which each cell decides its own sexual fate based on its sex chromosome constitution, but, surprisingly, recent data demonstrate that in Drosophila sex determination is not cell-autonomous, as it happens in mammals. Thus the presence of signals coordinating the development of a gender-specific phenotype cannot be excluded.This could explain why Wolbachia interferes with insect reproduction; and also could explain why Wolbachia interferes with insect development.Thus, is "sex (=reproduction) and stripping (=ecdysis)" the key to the intimate relationship between Wolbachia and its host?
Collapse
|
33
|
Okegbe TC, DiNardo S. The endoderm specifies the mesodermal niche for the germline in Drosophila via Delta-Notch signaling. Development 2011; 138:1259-67. [PMID: 21350008 DOI: 10.1242/dev.056994] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Interactions between niche cells and stem cells are vital for proper control over stem cell self-renewal and differentiation. However, there are few tissues where the initial establishment of a niche has been studied. The Drosophila testis houses two stem cell populations, which each lie adjacent to somatic niche cells. Although these niche cells sustain spermatogenesis throughout life, it is not understood how their fate is established. Here, we show that Notch signaling is necessary to specify niche cell fate in the developing gonad. Surprisingly, our results indicate that adjacent endoderm is the source of the Notch-activating ligand Delta. We also find that niche cell specification occurs earlier than anticipated, well before the expression of extant markers for niche cell fate. This work further suggests that endoderm plays a dual role in germline development. The endoderm assists both in delivering germ cells to the somatic gonadal mesoderm, and in specifying the niche where these cells will subsequently develop as stem cells. Because in mammals primordial germ cells also track through endoderm on their way to the genital ridge, our work raises the possibility that conserved mechanisms are employed to regulate germline niche formation.
Collapse
Affiliation(s)
- Tishina C Okegbe
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | |
Collapse
|
34
|
Negri I. Wolbachia as an "infectious" extrinsic factor manipulating host signaling pathways. Front Endocrinol (Lausanne) 2011; 2:115. [PMID: 22654845 PMCID: PMC3356060 DOI: 10.3389/fendo.2011.00115] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 12/21/2011] [Indexed: 11/13/2022] Open
Abstract
Wolbachia pipientis is a widespread endosymbiont of filarial nematodes and arthropods. While in worms the symbiosis is obligate, in arthropods Wolbachia induces several reproductive manipulations (i.e., cytoplasmic incompatibility, parthenogenesis, feminization of genetic males, and male-killing) in order to increase the number of infected females. These various phenotypic effects may be linked to differences in host physiology, and in particular to endocrine-related processes governing growth, development, and reproduction. Indeed, a number of evidences links Wolbachia symbiosis to insulin and ecdysteroid signaling, two multilayered pathways known to work antagonistically, jointly or even independently for the regulation of different molecular networks. At present it is not clear whether Wolbachia manipulates one pathway, thus affecting other related metabolic networks, or if it targets both pathways, even interacting at several points in each of them. Interestingly, in view of the interplay between hormone signaling and epigenetic machinery, a direct influence of the "infection" on hormonal signaling involving ecdysteroids might be achievable through the manipulation of the host's epigenetic pathways.
Collapse
Affiliation(s)
- Ilaria Negri
- Department of Exploitation and Protection of the Agricultural and Forestry Resources, Università di TorinoGrugliasco, Italy
- Koiné – Environmental ConsultingParma, Italy
- *Correspondence: Ilaria Negri, Department of Exploitation and Protection of the Agricultural and Forestry Resources, Università di Torino, Via L. da Vinci, 44, 10095 Grugliasco, Italy. e-mail:
| |
Collapse
|
35
|
Murray SM, Yang SY, Van Doren M. Germ cell sex determination: a collaboration between soma and germline. Curr Opin Cell Biol 2010; 22:722-9. [PMID: 21030233 PMCID: PMC2996260 DOI: 10.1016/j.ceb.2010.09.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 09/27/2010] [Accepted: 09/27/2010] [Indexed: 01/28/2023]
Abstract
Sex determination is regulated very differently in the soma vs. the germline, yet both processes are critical for the creation of the male and female gametes. In general, the soma plays an essential role in regulating sexual identity of the germline. However, in some species, such as Drosophila and mouse, the sex chromosome constitution of the germ cells makes an autonomous contribution to germline sexual development. Here we review how the soma and germline cooperate to determine germline sexual identity for some important model systems, the fly, the worm and the mouse, and discuss some of the implications of 'dual control' (soma plus germline) as compared to species where germline sex is dictated only by the surrounding soma.
Collapse
Affiliation(s)
| | | | - Mark Van Doren
- Department of Biology, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| |
Collapse
|
36
|
Murat S, Hopfen C, McGregor AP. The function and evolution of Wnt genes in arthropods. ARTHROPOD STRUCTURE & DEVELOPMENT 2010; 39:446-452. [PMID: 20685345 DOI: 10.1016/j.asd.2010.05.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 05/19/2010] [Accepted: 05/26/2010] [Indexed: 05/29/2023]
Abstract
Wnt signalling is required for a wide range of developmental processes, from cleavage to patterning and cell migration. There are 13 subfamilies of Wnt ligand genes and this diverse repertoire appeared very early in metazoan evolution. In this review, we first summarise the known Wnt gene repertoire in various arthropods. Insects appear to have lost several Wnt subfamilies, either generally, such as Wnt3, or in lineage specific patterns, for example, the loss of Wnt7 in Anopheles. In Drosophila and Acyrthosiphon, only seven and six Wnt subfamilies are represented, respectively; however, the finding of nine Wnt genes in Tribolium suggests that arthropods had a larger repertoire ancestrally. We then discuss what is currently known about the expression and developmental function of Wnt ligands in Drosophila and other insects in comparison to other arthropods, such as the spiders Achaearanea and Cupiennius. We conclude that studies of Wnt genes have given us much insight into the developmental roles of some of these ligands. However, given the frequent loss of Wnt genes in insects and the derived development of Drosophila, further studies of these important genes are required in a broader range of arthropods to fully understand their developmental function and evolution.
Collapse
Affiliation(s)
- Sophie Murat
- Institut für Populationsgenetik, Veterinärmedizinische Universität Wien, Veterinärplatz 1, Vienna, Austria
| | | | | |
Collapse
|
37
|
Callaghan TR, Degnan BM, Sellars MJ. Expression of sex and reproduction-related genes in Marsupenaeus japonicus. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2010; 12:664-677. [PMID: 20091331 DOI: 10.1007/s10126-009-9254-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2009] [Accepted: 12/11/2009] [Indexed: 05/28/2023]
Abstract
Expressed sequence tags (ESTs) were identified from reciprocal suppression subtractive hybridization cDNA libraries from Marsupenaeus japonicus (Kuruma shrimp) female and male gonads. The expression profiles of 24 of these ESTs were determined in female and male gonads and developing postlarvae by real-time quantitative reverse transcription-polymerase chain reaction. When expression was determined in gonads, six of the ESTs were expressed in ovaries only, and five of the ESTs were expressed in testes only. When expression was determined in whole individuals during postlarval development, expression of the ESTs was low and inconsistent until stage PL110 (110 days since metamorphosis from mysis stage to the first postlarval stage). At PL110, seven of the ESTs were detected in females only, and seven ESTs were detected in males only. Sex-specific expression at this developmental stage indicates that these ESTs act as important gonadal development markers and may have a role in gametogenesis.
Collapse
Affiliation(s)
- Tamera R Callaghan
- CSIRO Food Futures National Research Flagship, 5 Julius Avenue, North Ryde, Sydney, New South Wales 2113, Australia.
| | | | | |
Collapse
|
38
|
Cossais F, Sock E, Hornig J, Schreiner S, Kellerer S, Bösl MR, Russell S, Wegner M. Replacement of mouse Sox10 by the Drosophila ortholog Sox100B provides evidence for co-option of SoxE proteins into vertebrate-specific gene-regulatory networks through altered expression. Dev Biol 2010; 341:267-81. [PMID: 20144603 DOI: 10.1016/j.ydbio.2010.01.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 01/29/2010] [Accepted: 01/29/2010] [Indexed: 11/26/2022]
Abstract
Neural crest cells and oligodendrocytes as the myelinating glia of the central nervous system exist only in vertebrates. Their development is regulated by complex regulatory networks, of which the SoxE-type high-mobility-group domain transcription factors Sox8, Sox9 and Sox10 are essential components. Here we analyzed by in ovo electroporation in chicken and by gene replacement in the mouse whether the Drosophila ortholog Sox100B can functionally substitute for vertebrate SoxE proteins. Sox100B overexpression in the chicken neural tube led to the induction of neural crest cells as previously observed for vertebrate SoxE proteins. Furthermore, many aspects of neural crest and oligodendrocyte development were surprisingly normal in mice in which the Sox10 coding information was replaced by Sox100B arguing that Sox100B integrates well into the gene-regulatory networks that drive these processes. Our results therefore provide strong evidence for a model in which SoxE proteins were co-opted to these gene-regulatory networks mainly through the acquisition of novel expression patterns. However, later developmental defects in several neural crest derived lineages in mice homozygous for the Sox100B replacement allele indicate that some degree of functional specialization and adaptation of SoxE protein properties have taken place in addition to the co-option event.
Collapse
Affiliation(s)
- François Cossais
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen, Fahrstrasse 17, D-91054 Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Functional genomic identification of genes required for male gonadal differentiation in Caenorhabditis elegans. Genetics 2010; 185:523-35. [PMID: 20308279 DOI: 10.1534/genetics.110.116038] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Caenorhabditis elegans somatic gonad develops from a four-cell primordium into a mature organ that differs dramatically between the sexes in overall morphology (two arms in hermaphrodites and one in males) and in the cell types comprising it. Gonadal development in C. elegans is well studied, but regulation of sexual differentiation, especially later in gonadal development, remains poorly elucidated. To identify genes involved in this process, we performed a genome-wide RNAi screen using sex-specifically expressed gonadal GFP reporters. This screen identified several phenotypic classes, including approximately 70 genes whose depletion feminized male gonadal cells. Among the genes required for male cell fate specification are Wnt/beta-catenin pathway members, cell cycle regulators, and genes required for mitotic spindle function and cytokinesis. We find that a Wnt/beta-catenin pathway independent of extracellular Wnt ligand is essential for asymmetric cell divisions and male differentiation during gonadal development in larvae. We also find that the cell cycle regulators cdk-1 and cyb-3 and the spindle/cytokinesis regulator zen-4 are required for Wnt/beta-catenin pathway activity in the developing gonad. After sex is determined in the gonadal primordium the global sex determination pathway is dispensable for gonadal sexual fate, suggesting that male cell fates are promoted and maintained independently of the global pathway during this period.
Collapse
|
40
|
|
41
|
Casper AL, Van Doren M. The establishment of sexual identity in the Drosophila germline. Development 2009; 136:3821-30. [PMID: 19855024 PMCID: PMC2766343 DOI: 10.1242/dev.042374] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/14/2009] [Indexed: 12/11/2022]
Abstract
The establishment of sexual identity is a crucial step of germ cell development in sexually reproducing organisms. Sex determination in the germline is controlled differently than in the soma, and often depends on communication from the soma. To investigate how sexual identity is established in the Drosophila germline, we first conducted a molecular screen for genes expressed in a sex-specific manner in embryonic germ cells. Sex-specific expression of these genes is initiated at the time of gonad formation (stage 15), indicating that sexual identity in the germline is established by this time. Experiments where the sex of the soma was altered relative to that of the germline (by manipulating transformer) reveal a dominant role for the soma in regulating initial germline sexual identity. Germ cells largely take on the sex of the surrounding soma, although the sex chromosome constitution of the germ cells still plays some role at this time. The male soma signals to the germline through the JAK/STAT pathway, while the nature of the signal from the female soma remains unknown. We also find that the genes ovo and ovarian tumor (otu) are expressed in a female-specific manner in embryonic germ cells, consistent with their role in promoting female germline identity. However, removing the function of ovo and otu, or reducing germline function of Sex lethal, had little effect on establishment of germline sexual identity. This is consistent with our findings that signals from the soma are dominant over germline autonomous cues at the initial stage of germline sex determination.
Collapse
Affiliation(s)
- Abbie L Casper
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | |
Collapse
|
42
|
Sheng XR, Posenau T, Gumulak-Smith JJ, Matunis E, Van Doren M, Wawersik M. Jak-STAT regulation of male germline stem cell establishment during Drosophila embryogenesis. Dev Biol 2009; 334:335-44. [PMID: 19643104 PMCID: PMC2777977 DOI: 10.1016/j.ydbio.2009.07.031] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 07/09/2009] [Accepted: 07/10/2009] [Indexed: 11/16/2022]
Abstract
Germline stem cells (GSCs) in Drosophila are descendants of primordial germ cells (PGCs) specified during embryogenesis. The precise timing of GSC establishment in the testis has not been determined, nor is it known whether mechanisms that control GSC maintenance in the adult are involved in GSC establishment. Here, we determine that PGCs in the developing male gonad first become GSCs at the embryo to larval transition. This coincides with formation of the embryonic hub; the critical signaling center that regulates adult GSC behavior within the stem cell microenvironment (niche). We find that the Jak-STAT signaling pathway is activated in a subset of PGCs that associate with the newly-formed embryonic hub. These PGCs express GSC markers and function like GSCs, while PGCs that do not associate with the hub begin to differentiate. In the absence of Jak-STAT activation, PGCs adjacent to the hub fail to exhibit the characteristics of GSCs, while ectopic activation of the Jak-STAT pathway prevents differentiation. These findings show that stem cell formation is closely linked to development of the stem cell niche, and suggest that Jak-STAT signaling is required for initial establishment of the GSC population in developing testes.
Collapse
Affiliation(s)
- X. Rebecca Sheng
- Department of Cell Biology, 725 N. Wolfe Street, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Trevor Posenau
- Department of Biology, Integrated Science Center, Rm 2139 The College of William and Mary, Williamsburg, VA, 23185, USA
| | - Juliann J. Gumulak-Smith
- Department of Biology, Integrated Science Center, Rm 2139 The College of William and Mary, Williamsburg, VA, 23185, USA
| | - Erika Matunis
- Department of Cell Biology, 725 N. Wolfe Street, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mark Van Doren
- Department of Biology, Mudd Hall 305, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Matthew Wawersik
- Department of Biology, Integrated Science Center, Rm 2139 The College of William and Mary, Williamsburg, VA, 23185, USA
- Department of Biology, Mudd Hall 305, The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| |
Collapse
|
43
|
Chau J, Kulnane LS, Salz HK. Sex-lethal facilitates the transition from germline stem cell to committed daughter cell in the Drosophila ovary. Genetics 2009; 182:121-32. [PMID: 19237687 PMCID: PMC2674811 DOI: 10.1534/genetics.109.100693] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2009] [Accepted: 02/19/2009] [Indexed: 11/18/2022] Open
Abstract
In Drosophila, the female-specific SEX-LETHAL (SXL) protein is required for oogenesis, but how Sxl interfaces with the genetic circuitry controlling oogenesis remains unknown. Here we use an allele of sans fille (snf) that specifically eliminates SXL protein in germ cells to carry out a detailed genetic and cell biological analysis of the resulting ovarian tumor phenotype. We find that tumor growth requires both Cyclin B and zero population growth, demonstrating that these mutant cells retain at least some of the essential growth-control mechanisms used by wild-type germ cells. Using a series of molecular markers, we establish that while the tumor often contains at least one apparently bona fide germline stem cell, the majority of cells exhibit an intermediate fate between a stem cell and its daughter cell fated to differentiate. In addition, snf tumors misexpress a select group of testis-enriched markers, which, remarkably, are also misexpressed in ovarian tumors that arise from the loss of bag of marbles (bam). Results of genetic epistasis experiments further reveal that bam's differentiation-promoting function depends on Sxl. Together these data demonstrate a novel role for Sxl in the lineage progression from stem cell to committed daughter cell and suggest a model in which Sxl partners with bam to facilitate this transition.
Collapse
Affiliation(s)
- Johnnie Chau
- Department of Genetics, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106-4955, USA
| | | | | |
Collapse
|
44
|
Nanda S, DeFalco T, Hui Yong Loh S, Phochanukul N, Camara N, Van Doren M, Russell S. Sox100B, a Drosophila group E Sox-domain gene, is required for somatic testis differentiation. Sex Dev 2009; 3:26-37. [PMID: 19339815 PMCID: PMC2836946 DOI: 10.1159/000200079] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Accepted: 11/26/2008] [Indexed: 11/19/2022] Open
Abstract
Sex determination mechanisms are thought to evolve rapidly and show little conservation among different animal species. For example, the critical gene on the Y chromosome, SRY, that determines sex in most mammals, is not found in other animals. However, a related Sox domain transcription factor, SOX9, is also required for testis development in mammals and exhibits male-specific gonad expression in other vertebrate species. Previously, we found that the Drosophila orthologue of SOX9, Sox100B, is expressed male-specifically during gonad development. We now investigate the function of Sox100B and find, strikingly, that Sox100B is essential for testis development in Drosophila. In Sox100B mutants, the adult testis is severely reduced and fails to interact with other parts of the reproductive tract, which are themselves unaffected. While a testis initially forms in Sox100B mutants, it fails to undergo proper morphogenesis during pupal stages, likely due to defects in the pigment cells. In contrast, no substantive defects are observed in ovary development in Sox100B mutant females. Thus, as is observed in mammals, a Sox9 homolog is essential for sex-specific gonad development in Drosophila, suggesting that the molecular mechanisms regulating sexually dimorphic gonad development may be more conserved than previously suspected.
Collapse
Affiliation(s)
- S. Nanda
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - T.J. DeFalco
- Department of Biology, Johns Hopkins University, Baltimore, Md., USA
| | - S. Hui Yong Loh
- Department of Genetics, University of Cambridge, Cambridge, UK
- MRC Toxicology Unit, Hodgkin Building, University of Leicester, Leicester, UK
| | - N. Phochanukul
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - N. Camara
- Department of Biology, Johns Hopkins University, Baltimore, Md., USA
| | - M. Van Doren
- Department of Biology, Johns Hopkins University, Baltimore, Md., USA
| | - S. Russell
- Department of Genetics, University of Cambridge, Cambridge, UK
| |
Collapse
|
45
|
Sexual back talk with evolutionary implications: stimulation of the Drosophila sex-determination gene sex-lethal by its target transformer. Genetics 2008; 180:1963-81. [PMID: 18845845 DOI: 10.1534/genetics.108.093898] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
We describe a surprising new regulatory relationship between two key genes of the Drosophila sex-determination gene hierarchy, Sex-lethal (Sxl) and transformer (tra). A positive autoregulatory feedback loop for Sxl was known to maintain somatic cell female identity by producing SXL-F protein to continually instruct the target gene transformer (tra) to make its feminizing product, TRA-F. We discovered the reciprocal regulatory effect by studying genetically sensitized females: TRA-F from either maternal or zygotic tra expression stimulates Sxl-positive autoregulation. We found female-specific tra mRNA in eggs as predicted by this tra maternal effect, but not predicted by the prevailing view that tra has no germline function. TRA-F stimulation of Sxl seems to be direct at some point, since Sxl harbors highly conserved predicted TRA-F binding sites. Nevertheless, TRA-F stimulation of Sxl autoregulation in the gonadal soma also appears to have a cell-nonautonomous aspect, unprecedented for somatic Sxl regulation. This tra-Sxl retrograde regulatory circuit has evolutionary implications. In some Diptera, tra occupies Sxl's position as the gene that epigenetically maintains female identity through direct positive feedback on pre-mRNA splicing. The tra-mediated Sxl feedback in Drosophila may be a vestige of regulatory redundancy that facilitated the evolutionary transition from tra to Sxl as the master sex switch.
Collapse
|
46
|
Camara N, Whitworth C, Van Doren M. The creation of sexual dimorphism in the Drosophila soma. Curr Top Dev Biol 2008; 83:65-107. [PMID: 19118664 DOI: 10.1016/s0070-2153(08)00403-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Animals have evolved a fascinating array of mechanisms for conducting sexual reproduction. These include producing the sex-specific gametes, as well as mechanisms for attracting a mate, courting a mate, and getting the gametes together. These processes require that males and females take on dramatically different forms (sexual dimorphism). Here, we will explore the problem of how sex is determined in Drosophila, and pay particular attention to how information about sexual identity is used to instruct males and females to develop differently. Along the way, we will highlight new work that challenges some of the traditional views about sex determination. In Drosophila, it is commonly thought that every cell decides its own sex based on its sex chromosome constitution (XX vs. XY). However, we now know that many cell types undergo nonautonomous sex determination, where they are told what sex to be through signals from surrounding cells, independent of their own chromosomal content. Further, it now appears that not all cells even "know" their sex, since key members of the sex determination pathway are not expressed in all cells. Thus, our understanding of how sex is determined, and how sexual identity is used to create sexual dimorphism, has changed considerably.
Collapse
Affiliation(s)
- Nicole Camara
- Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | | | | |
Collapse
|