1
|
Walker BM, Palumbo RJ, Knutson BA. Tissue-specific requirement of Polr1D in the prothoracic gland for ecdysone-mediated developmental transitions in Drosophila melanogaster. Dev Dyn 2025. [PMID: 40317818 DOI: 10.1002/dvdy.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND POLR1D is a shared subunit of RNA Polymerases I and III, which transcribe the rRNA incorporated into ribosomes. Mutations in POLR1D cause Treacher Collins syndrome, a craniofacial disorder that arises from impaired ribosome biogenesis in neural crest cells. Previously, we found that RNAi knockdown of Polr1D in several non-neural Drosophila tissues caused developmental defects that phenocopy mutations affecting ecdysone signaling. Ecdysone is a steroid hormone produced in the prothoracic gland (PG) of insects that triggers developmental transitions. Here, we show that Polr1D is required for PG development and ecdysone production to facilitate larval developmental transitions. RESULTS We found that Polr1D RNAi in the PG causes larval developmental arrest due to defective peripheral ecdysone signaling. We also found that Polr1D is required for the growth of PG cells and for maintaining nucleolar structure. We found that Polr1D is required for the synthesis of mature ribosomes and the production of the Pol III-transcribed 7SK RNA. Furthermore, developmental arrest of Polr1D RNAi larvae and Polr1D mutant (G30R) larvae was partially rescued by treatment with exogenous ecdysone. CONCLUSION These results demonstrate a role for Drosophila Polr1D in PG development and suggest that disruptions in human Polr1D might impact additional cell types during development.
Collapse
Affiliation(s)
- Bridget M Walker
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Ryan J Palumbo
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Bruce A Knutson
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
2
|
Harsh S, Liu HY, Bhaskar PK, Rushlow C, Bach EA. The pioneer factor Zelda induces male-to-female somatic sex reversal in adult tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645575. [PMID: 40236223 PMCID: PMC11996320 DOI: 10.1101/2025.03.26.645575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Somatic sex identity must be maintained throughout adulthood for tissue function. Adult somatic stem cells in the Drosophila testis (i.e., CySCs) lacking the transcription factor Chinmo are reprogrammed to their ovarian counterparts by induction of female-specific Tra F , but this is not mechanistically understood. Pioneer factors play central roles in direct reprogramming, and many upregulated genes in chinmo -/- CySCs contain binding sites for the pioneer factor Zelda (Zld). microRNAs repress zld mRNA in wild type CySCs, but they are downregulated after Chinmo loss, allowing for zld mRNA translation. Zld depletion from chinmo -/- CySCs suppresses feminization, and ectopic Zld induces Tra F and feminizes wild-type CySCs. qkr58E-2 and ecdysone receptor ( EcR ), direct Zld targets in the embryo, are female-biased in adult gonads and upregulated in chinmo -/- CySCs. The RNA-binding protein Qkr58E-2 produces Tra F , while EcR promotes female-biased gene expression. Ectopic Zld feminizes adult male adipose tissue, demonstrating that Zld can instruct female and override male identity in adult XY tissues. Highlights zld mRNA is repressed by microRNAs in XY somatic gonadal cells Zld is upregulated in and required for sex reversal of XY chinmo -/- cells Zld induces Qkr58E-2 and EcR, which cause Tra F and female-biased transcription Zld feminizes XY adipose cells by inducing Tra F and downregulating Chinmo.
Collapse
|
3
|
Kwon SY, Chan K, Stofanko M, Chan KH, Badenhorst P. Abrupt-mediated control of ninjurins regulates Drosophila sessile haemocyte compartments. Development 2024; 151:dev202977. [PMID: 39545919 DOI: 10.1242/dev.202977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024]
Abstract
Macrophage-like cells called haemocytes are key effectors of Drosophila cellular innate immune function. Larval haemocytes exist either in circulation or localize to segmentally repeated sessile haemocyte compartments (SHCs). While numerous functions have been proposed for SHCs, the mechanisms directing haemocytes to them are unclear. Here, we have exploited the developmentally regulated dispersal of SHCs that occurs at pupariation to identify the Abrupt (Ab) transcription factor (TF) and ninjurin cell-adhesion molecules as regulators of haemocyte recruitment to SHCs. We show that larval haemocytes express ninjurins, which are required for targeting haemocytes to SHCs. However, at pupariation, ecdysteroid signalling stimulates Ab expression, which collaborates with TFs, including Blimp-1 and Hr3, to repress ninjurins and disperse haemocytes. We observe that experimental manipulations that antagonize ninjurin function in larval haemocytes cause premature SHC dispersal, while stabilization of ninjurins in haemocytes blocks developmentally regulated SHC remodelling and increases sensitivity to immune challenges. Cumulatively, our data indicate that control of ninjurin activity provides a common target through which diverse developmental, environmental and immune stimuli can be integrated to control haemocyte dispersal and immune function.
Collapse
Affiliation(s)
- So Yeon Kwon
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Kimberly Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Martin Stofanko
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Ka Hei Chan
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| | - Paul Badenhorst
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston B15 2TT, UK
| |
Collapse
|
4
|
Morrow H, Mirth CK. Timing Drosophila development through steroid hormone action. Curr Opin Genet Dev 2024; 84:102148. [PMID: 38271845 DOI: 10.1016/j.gde.2023.102148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 01/27/2024]
Abstract
Specifically timed pulses of the moulting hormone ecdysone are necessary for developmental progression in insects, guiding development through important milestones such as larval moults, pupation and metamorphosis. It also coordinates the acquisition of cell identities, known as cell patterning, and growth in a tissue-specific manner. In the absence of ecdysone, the ecdysone receptor heterodimer Ecdysone Receptor and Ultraspiracle represses expression of target primary response genes, which become de-repressed as the ecdysone titre rises. However, ecdysone signalling elicits both repressive and activating responses in a temporal and tissue-specific manner. To understand how ecdysone achieves such specificity, this review explores the layers of gene regulation involved in stage-appropriate ecdysone responses in Drosophila fruit flies.
Collapse
Affiliation(s)
- Hannah Morrow
- School of Biological Sciences, Monash University, Clayton, Victoria 3000, Australia.
| | - Christen K Mirth
- School of Biological Sciences, Monash University, Clayton, Victoria 3000, Australia
| |
Collapse
|
5
|
Suyama R, Cetraro N, Yew JY, Kai T. Microbes control Drosophila germline stem cell increase and egg maturation through hormonal pathways. Commun Biol 2023; 6:1287. [PMID: 38123715 PMCID: PMC10733356 DOI: 10.1038/s42003-023-05660-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023] Open
Abstract
Reproduction is highly dependent on environmental and physiological factors including nutrition, mating stimuli and microbes. Among these factors, microbes facilitate vital functions for host animals such as nutritional intake, metabolic regulation, and enhancing fertility under poor nutrition conditions. However, detailed molecular mechanisms by which microbes control germline maturation, leading to reproduction, remain largely unknown. In this study, we show that environmental microbes exert a beneficial effect on Drosophila oogenesis by promoting germline stem cell (GSC) proliferation and subsequent egg maturation via acceleration of ovarian cell division and suppression of apoptosis. Moreover, insulin-related signaling is not required; rather, the ecdysone pathway is necessary for microbe-induced increase of GSCs and promotion of egg maturation, while juvenile hormone contributes only to increasing GSC numbers, suggesting that hormonal pathways are activated at different stages of oogenesis. Our findings reveal that environmental microbes can enhance host reproductivity by modulating host hormone release and promoting oogenesis.
Collapse
Affiliation(s)
- Ritsuko Suyama
- Laboratory of Germline Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka Suita, Osaka, 565-0871, Japan.
| | - Nicolas Cetraro
- Pacific Biosciences Research Center, University of Hawai'i at Manoa, 1993 East-West Road, Honolulu, HI, 96822, USA
| | - Joanne Y Yew
- Pacific Biosciences Research Center, University of Hawai'i at Manoa, 1993 East-West Road, Honolulu, HI, 96822, USA.
| | - Toshie Kai
- Laboratory of Germline Biology, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
6
|
Godneeva B, Ninova M, Fejes-Toth K, Aravin A. SUMOylation of Bonus, the Drosophila homolog of Transcription Intermediary Factor 1, safeguards germline identity by recruiting repressive chromatin complexes to silence tissue-specific genes. eLife 2023; 12:RP89493. [PMID: 37999956 PMCID: PMC10672805 DOI: 10.7554/elife.89493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023] Open
Abstract
The conserved family of Transcription Intermediary Factors (TIF1) proteins consists of key transcriptional regulators that control transcription of target genes by modulating chromatin state. Unlike mammals that have four TIF1 members, Drosophila only encodes one member of the family, Bonus. Bonus has been implicated in embryonic development and organogenesis and shown to regulate several signaling pathways, however, its targets and mechanism of action remained poorly understood. We found that knockdown of Bonus in early oogenesis results in severe defects in ovarian development and in ectopic expression of genes that are normally repressed in the germline, demonstrating its essential function in the ovary. Recruitment of Bonus to chromatin leads to silencing associated with accumulation of the repressive H3K9me3 mark. We show that Bonus associates with the histone methyltransferase SetDB1 and the chromatin remodeler NuRD and depletion of either component releases Bonus-induced repression. We further established that Bonus is SUMOylated at a single site at its N-terminus that is conserved among insects and this modification is indispensable for Bonus's repressive activity. SUMOylation influences Bonus's subnuclear localization, its association with chromatin and interaction with SetDB1. Finally, we showed that Bonus SUMOylation is mediated by the SUMO E3-ligase Su(var)2-10, revealing that although SUMOylation of TIF1 proteins is conserved between insects and mammals, both the mechanism and specific site of modification is different in the two taxa. Together, our work identified Bonus as a regulator of tissue-specific gene expression and revealed the importance of SUMOylation as a regulator of complex formation in the context of transcriptional repression.
Collapse
Affiliation(s)
- Baira Godneeva
- California Institute of Technology, Division of Biology and Biological EngineeringPasadenaUnited States
- Institute of Gene Biology, Russian Academy of SciencesMoscowRussian Federation
| | - Maria Ninova
- University of California, RiversideRiversideUnited States
| | - Katalin Fejes-Toth
- California Institute of Technology, Division of Biology and Biological EngineeringPasadenaUnited States
| | - Alexei Aravin
- California Institute of Technology, Division of Biology and Biological EngineeringPasadenaUnited States
| |
Collapse
|
7
|
Singh M, Kumar S. Effect of single nucleotide polymorphisms on the structure of long noncoding RNAs and their interaction with RNA binding proteins. Biosystems 2023; 233:105021. [PMID: 37703988 DOI: 10.1016/j.biosystems.2023.105021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/25/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Long non-coding RNAs (lncRNA) are emerging as a new class of regulatory RNAs with remarkable potential to be utilized as therapeutic targets against many human diseases. Several genome-wide association studies (GWAS) have catalogued Single Nucleotide Polymorphisms (SNPs) present in the noncoding regions of the genome from where lncRNAs originate. In this study, we have selected 67 lncRNAs with GWAS-tagged SNPs and have also investigated their role in affecting the local secondary structures. Majority of the SNPs lead to changes in the secondary structure of lncRNAs to a different extent by altering the base pairing patterns. These structural changes in lncRNA are also manifested in form of alteration in the binding site for RNA binding proteins (RBPs) along with affecting their binding efficacies. Ultimately, these structural modifications may influence the transcriptional and post-transcriptional pathways of these RNAs, leading to the causation of diseases. Hence, it is important to understand the possible underlying mechanism of RBPs in association with GWAS-tagged SNPs in human diseases.
Collapse
Affiliation(s)
- Mandakini Singh
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India
| | - Santosh Kumar
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, 769008, India.
| |
Collapse
|
8
|
Sun Z, Nystul TG, Zhong G. Single-cell RNA sequencing identifies eggplant as a regulator of germ cell development in Drosophila. EMBO Rep 2023; 24:e56475. [PMID: 37603128 PMCID: PMC10561367 DOI: 10.15252/embr.202256475] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023] Open
Abstract
Drosophila ovarian germline stem cells (GSCs) are a powerful model for stem cell research. In this study, we use single-cell RNA sequencing (scRNA-seq), an RNAi screen and bioinformatic analysis, to identify genes involved in germ cell differentiation, including 34 genes with upregulated expression during early germ cell development and 19 genes that may regulate germ cell differentiation. Among these, a gene we have named eggplant (eggpl) is highly expressed in GSCs and downregulated in early daughter cells. RNAi knockdown of eggpl causes germ cell proliferation and differentiation defects. In flies fed a rich yeast diet, the expression of eggpl is significantly lower and knockdown or knockout of eggpl phenocopies a rich diet. In addition, eggpl knockdown suppresses the reduction in germ cell proliferation caused by inhibition of the insulin effector PI3K. These findings suggest that downregulation of eggpl links nutritional status to germ cell proliferation and differentiation. Collectively, this study provides new insights into the signaling networks that regulate early germ cell development and identifies eggpl as a key player in this process.
Collapse
Affiliation(s)
- Zhipeng Sun
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture and Rural AffairsSouth China Agricultural UniversityGuangzhouChina
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of EducationSouth China Agricultural UniversityGuangzhouChina
| | | | - Guohua Zhong
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture and Rural AffairsSouth China Agricultural UniversityGuangzhouChina
- Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of EducationSouth China Agricultural UniversityGuangzhouChina
| |
Collapse
|
9
|
Godneeva B, Ninova M, Fejes Tóth K, Aravin AA. SUMOylation of Bonus, the Drosophila homolog of Transcription Intermediary Factor 1, safeguards germline identity by recruiting repressive chromatin complexes to silence tissue-specific genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.14.536936. [PMID: 37645991 PMCID: PMC10461926 DOI: 10.1101/2023.04.14.536936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The conserved family of Transcription Intermediary Factors (TIF1) proteins consists of key transcriptional regulators that control transcription of target genes by modulating chromatin state. Unlike mammals that have four TIF1 members, Drosophila only encodes one member of the family, Bonus. Bonus has been implicated in embryonic development and organogenesis and shown to regulate several signaling pathways, however, its targets and mechanism of action remained poorly understood. We found that knockdown of Bonus in early oogenesis results in severe defects in ovarian development and in ectopic expression of genes that are normally repressed in the germline, demonstrating its essential function in the ovary. Recruitment of Bonus to chromatin leads to silencing associated with accumulation of the repressive H3K9me3 mark. We show that Bonus associates with the histone methyltransferase SetDB1 and the chromatin remodeler NuRD and depletion of either component releases Bonus-induced repression. We further established that Bonus is SUMOylated at a single site at its N-terminus that is conserved among insects and this modification is indispensable for Bonus's repressive activity. SUMOylation influences Bonus's subnuclear localization, its association with chromatin and interaction with SetDB1. Finally, we showed that Bonus SUMOylation is mediated by the SUMO E3-ligase Su(var)2-10, revealing that although SUMOylation of TIF1 proteins is conserved between insects and mammals, both the mechanism and specific site of modification is different in the two taxa. Together, our work identified Bonus as a regulator of tissue-specific gene expression and revealed the importance of SUMOylation as a regulator of complex formation in the context of transcriptional repression.
Collapse
Affiliation(s)
- Baira Godneeva
- California Institute of Technology, Division of Biology and Biological Engineering, Pasadena, CA 91125, USA
- Institute of Gene Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Maria Ninova
- University of California, Riverside, Riverside, CA 92521, USA
| | - Katalin Fejes Tóth
- California Institute of Technology, Division of Biology and Biological Engineering, Pasadena, CA 91125, USA
| | - Alexei A. Aravin
- California Institute of Technology, Division of Biology and Biological Engineering, Pasadena, CA 91125, USA
| |
Collapse
|
10
|
Li T, Ye Y, Wu P, Luo R, Zhang H, Zheng W. Proteasome β3 subunit (PSMB3) controls female reproduction by promoting ecdysteroidogenesis during sexual maturation in Bactrocera dorsalis. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 157:103959. [PMID: 37172766 DOI: 10.1016/j.ibmb.2023.103959] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/06/2023] [Accepted: 05/08/2023] [Indexed: 05/15/2023]
Abstract
Steroid hormone 20-hydroxyecdysone (20E) plays critical roles in reproductive development in dipterans and several other insect species. Ecdysteroidogenesis in the glands of larval or nymphal insects and other arthropods has been extensively studied, but that in the adult gonads remains largely unknown. Here we identified a proteasome β3 subunit (PSMB3) from a highly invasive pest Bactrocera dorsalis, and found that this gene was crucial for ecdysone production during female reproduction. PSMB3 was enriched in the ovary, and it was upregulated during sexual maturation. RNAi-mediated depletion of PSMB3 resulted in retarded ovarian development and decreased fecundity. Additionally, knockdown of PSMB3 reduced 20E titer in hemolymph of B. dorsalis. Molecularly, RNA sequencing and qPCR validation revealed that PSMB3 depletion suppressed the expression of 20E biosynthetic genes in the ovary and 20E responsive genes in the ovary and fat body. Furthermore, exogenous 20E rescued the inhibition of the ovarian development caused by PSMB3 depletion. Taken together, this study provides new insights into the adult reproductive development-related biological processes controlled by PSMB3, and proposed a potential eco-friendly control strategy against this notorious agricultural pest.
Collapse
Affiliation(s)
- Tianran Li
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yinhao Ye
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Peng Wu
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Rengang Luo
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hongyu Zhang
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Weiwei Zheng
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China.
| |
Collapse
|
11
|
Rumbo M, Pagone V, Piulachs MD. Diverse functions of the ecdysone receptor (EcR) in the panoistic ovary of the German cockroach. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 156:103935. [PMID: 36996928 DOI: 10.1016/j.ibmb.2023.103935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 05/05/2023]
Abstract
Ecdysone regulates essential processes in insect life. Perhaps the most well-known of these are related to metamorphosis. However, ecdysone is also required to regulate the proliferation and differentiation of germ cells in the ovary. The role of ecdysone in insect oogenesis has been studied in depth in holometabolan species with meroistic ovaries, such as Drosophila melanogaster, while in hemimetabolan species with panoistic ovaries their functions are still poorly understood. In the present work, we studied the role of ecdysone in the ovary of the last nymphal instar of the cockroach Blattella germanica by using RNA interference to reduce the levels of the ecdysone receptor (EcR), and thereby deplete the expression of ecdysteroidogenic genes in the prothoracic gland. However, the expression of ecdysteroidogenic genes was upregulated in the ovary, resulting in cell overproliferation in the germarium, which appeared swollen. By analysing the expression of genes that respond to ecdysone, we found that when the source of 20E is the nymphal ovary, EcR appears to repress 20E-associated genes bypassing early genes signalling.
Collapse
Affiliation(s)
- M Rumbo
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain
| | - V Pagone
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain
| | - M D Piulachs
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain.
| |
Collapse
|
12
|
Soriano A, Petit C, Ryan S, Jemc JC. Tracking Follicle Cell Development. Methods Mol Biol 2023; 2626:151-177. [PMID: 36715904 DOI: 10.1007/978-1-0716-2970-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Somatic follicle cells are critical support cells for Drosophila oogenesis, as they provide signals and molecules needed to produce a mature egg. Throughout this process, the follicle cells differentiate into multiple subpopulations and transition between three different cell cycle programs to support nurse cell and oocyte development. The follicle cells are mitotic in early egg chamber development, as they cover the germline cyst. In mid-oogenesis, follicle cells switch from mitosis to endocycling, increasing their ploidy from 2C to 16C. Finally, in late oogenesis, cells transition from endocycling to gene amplification, increasing the copy number of a small subset of genes, including the genes encoding proteins required for egg maturation. In order to explore the genetic regulation of these cell cycle switches and follicle cell development and specification, clonal analysis and the GAL4/UAS system are used frequently to reduce or increase expression of genes of interest. These genetic approaches combined with immunohistochemistry and in situ hybridization are powerful tools for characterizing the mechanisms regulating follicle cell development and the mitosis/endocycle and endocycle/gene amplification transitions. This chapter describes the genetic tools available to manipulate gene expression in follicle cells, as well as the methods and reagents that can be utilized to explore gene expression throughout follicle cell development.
Collapse
Affiliation(s)
- Adrianna Soriano
- Department of Biology, Loyola University Chicago, Chicago, IL, USA.,Houston Baptist University, Houston, TX, USA
| | | | - Savannah Ryan
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Jennifer C Jemc
- Department of Biology, Loyola University Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Jin J, Zhao T. Niche formation and function in developing tissue: studies from the Drosophila ovary. Cell Commun Signal 2023; 21:23. [PMID: 36707894 PMCID: PMC9881360 DOI: 10.1186/s12964-022-01035-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/28/2022] [Indexed: 01/28/2023] Open
Abstract
Adult stem cells have a unique ability to self-renew and to generate differentiated daughter cells that are required in the body tissues. The identity of adult stem cells is maintained by extrinsic signals from other cell types, known as niche cells. Thus, the niche is required for appropriate tissue homeostasis. Niche is formed and recruits stem cells during tissue development; therefore, it is essential to establish niche cells and stem cells in proper numbers during development. A small niche may recruit too few stem cells and cause tissue degeneration, while a large niche may maintain too many stem cells and lead to tumorigenesis. Given that vertebrate tissues are not suitable for large-scale forward genetics studies, the Drosophila ovary stands out as an excellent model for studying how multiple niche cell types and germ cells (GCs) are coordinately regulated in vivo. Recent studies are beginning to reveal how various signaling molecules regulate niche formation and how niche cells non-autonomously influence GC number. In this review, we summarize the ovarian niche structure, the key signaling pathways for niche formation, and how niche cells generate extrinsic factors to control GC proliferation during ovarian development. Video abstract.
Collapse
Affiliation(s)
- Jian Jin
- grid.440646.40000 0004 1760 6105School of Educational Science, Anhui Normal University, Wuhu, 241000 People’s Republic of China
| | - Ting Zhao
- grid.411407.70000 0004 1760 2614School of Life Science, Hubei Key Laboratory of Genetic Regulation and Integrative Biology, Central China Normal University, Wuhan, 430079 People’s Republic of China
| |
Collapse
|
14
|
Khalid MZ, Sun Z, Zhang J, Zhang S, Zhong G. Cyromazine affects the ovarian germ cells of Drosophila via the ecdysone signaling pathway. Front Physiol 2022; 13:992306. [PMID: 36246127 PMCID: PMC9557234 DOI: 10.3389/fphys.2022.992306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Cyromazine, an insect growth regulator, has been extensively used against the insect pests of livestock and households. Previously, it was observed that the continuous selection of cyromazine from the larval to the adult stage decreased the number of germline stem cells (GSCs) and cystoblasts (CBs) in the adult ovary. In addition, in this study, we observed that the number of primordial germ cells (PGCs) was also decreased in the larval ovary after treatment with cyromazine. However, the mechanism by which it affects the germ cells is yet to be explored. Consequently, to deeply investigate the effects of cyromazine on the germ cells, we performed tissue-specific RNA sequencing. Bioinformatics analysis revealed that the ecdysone signaling pathway was significantly influenced under cyromazine stress. Based on that, we screened and selected 14 ecdysone signaling responsive genes and silenced their expression in the germ cells only. Results of that showed a considerable reduction in the number of germ cells. Furthermore, we mixed exogenous 20E with the cyromazine-containing diet to rescue the ecdysone signaling. Our results supported that the application of exogenous 20E significantly rescued the germ cells in the transgenic lines. Therefore, this implies that the cyromazine decreased the number of germ cells by affecting the ecdysone signaling pathway.
Collapse
|
15
|
Chen SL, Liu BT, Lee WP, Liao SB, Deng YB, Wu CL, Ho SM, Shen BX, Khoo GH, Shiu WC, Chang CH, Shih HW, Wen JK, Lan TH, Lin CC, Tsai YC, Tzeng HF, Fu TF. WAKE-mediated modulation of cVA perception via a hierarchical neuro-endocrine axis in Drosophila male-male courtship behaviour. Nat Commun 2022; 13:2518. [PMID: 35523813 PMCID: PMC9076693 DOI: 10.1038/s41467-022-30165-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/19/2022] [Indexed: 12/18/2022] Open
Abstract
The nervous and endocrine systems coordinate with each other to closely influence physiological and behavioural responses in animals. Here we show that WAKE (encoded by wide awake, also known as wake) modulates membrane levels of GABAA receptor Resistance to Dieldrin (Rdl), in insulin-producing cells of adult male Drosophila melanogaster. This results in changes to secretion of insulin-like peptides which is associated with changes in juvenile hormone biosynthesis in the corpus allatum, which in turn leads to a decrease in 20-hydroxyecdysone levels. A reduction in ecdysone signalling changes neural architecture and lowers the perception of the male-specific sex pheromone 11-cis-vaccenyl acetate by odorant receptor 67d olfactory neurons. These finding explain why WAKE-deficient in Drosophila elicits significant male-male courtship behaviour. The authors show that the Drosophila master regulator WAKE modulates the secretion of insulin-like peptides, triggering a decrease in 20-hydroxyecdysone levels. This lowers the perception of a male-specific sex pheromone and explains why WAKE-deficient Drosophila flies show male-male courtship behaviour.
Collapse
Affiliation(s)
- Shiu-Ling Chen
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Bo-Ting Liu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Wang-Pao Lee
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Sin-Bo Liao
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.,Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yao-Bang Deng
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Chia-Lin Wu
- Department of Biochemistry and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu, Taiwan
| | - Shuk-Man Ho
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Bing-Xian Shen
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Guan-Hock Khoo
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan
| | - Wei-Chiang Shiu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Chih-Hsuan Chang
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan.,Institute of Systems Neuroscience, National Tsing Hua University, Hsinchu, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Hui-Wen Shih
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan
| | - Jung-Kun Wen
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Tsuo-Hung Lan
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Tsaotun Psychiatric Center, Ministry of Health and Welfare, Nantou, Taiwan.,Department of Psychiatry, Taichung Veterans General Hospital, Taichung, Taiwan.,Center for Neuropsychiatric Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chih-Chien Lin
- Department of Psychiatry, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chen Tsai
- Department of Life Science and Life Science Center, Tunghai University, Taichung, Taiwan.
| | - Huey-Fen Tzeng
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| | - Tsai-Feng Fu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan.
| |
Collapse
|
16
|
Cyromazine Effects the Reproduction of Drosophila by Decreasing the Number of Germ Cells in the Female Adult Ovary. INSECTS 2022; 13:insects13050414. [PMID: 35621750 PMCID: PMC9144682 DOI: 10.3390/insects13050414] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 02/08/2023]
Abstract
Simple Summary Cyromazine, an insect growth regulator, is used to control the Dipteran pest population. Previous findings observed that treatment with cyromazine increased the larval mortality, by interfering with the ecdysone signaling. In addition, the application of exogenous 20E significantly reduced the mortality caused by cyromazine. Many studies have also supported the role of ecdysone signaling in the maintenance of germline stem cells (GSCs), where mutations in ecdysone signaling-related genes significantly decreased the number of GSCs. However, to date, no study has reported the effect of cyromazine on the GSCs of Drosophila melanogaster. In the present study, we observed that cyromazine significantly reduced the number of both GSCs and cystoblasts (CBs) in the ovary of adult female. To further understand the effect of cyromazine on germ cells, we selected some key genes related to the ecdysone signaling pathway and evaluated their expression through RT-qPCR. Additionally, we measured the ecdysone titer from the cyromazine-treated ovaries. Our results indicated a significant decrease in the expression of ecdysone signaling-related genes and also in the ecdysone titer. These results further supported our findings that cyromazine reduced the number of germ cells by interfering with the ecdysone signaling pathway. Abstract In the present study, we observed a 58% decrease in the fecundity of Drosophila melanogaster, after treatment with the cyromazine. To further elucidate the effects of cyromazine on reproduction, we counted the number of both germline stem cells (GSCs) and cystoblasts (CBs) in the ovary of a 3-day-old adult female. The results showed a significant decrease in the number of GSCs and CBs as compared to the control group. The mode of action of cyromazine is believed to be through the ecdysone signaling pathway. To further support this postulate, we observed the expression of key genes involved in the ecdysone signaling pathway and also determined the ecdysone titer from cyromazine-treated ovaries. Results indicated a significant decrease in the expression of ecdysone signaling-related genes as compared to the control group. Furthermore, the titer of the ecdysone hormone was also markedly reduced (90%) in cyromazine-treated adult ovaries, suggesting that ecdysone signaling was directly related to the decrease in the number of GSCs and CBs. However, further studies are required to understand the mechanism by which cyromazine affects the GSCs and CBs in female adult ovaries.
Collapse
|
17
|
Tarikere S, Ylla G, Extavour CG. Distinct gene expression dynamics in germ line and somatic tissue during ovariole morphogenesis in Drosophila melanogaster. G3 (BETHESDA, MD.) 2022; 12:jkab305. [PMID: 34849771 PMCID: PMC9210308 DOI: 10.1093/g3journal/jkab305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022]
Abstract
The survival and evolution of a species is a function of the number of offspring it can produce. In insects, the number of eggs that an ovary can produce is a major determinant of reproductive capacity. Insect ovaries are made up of tubular egg-producing subunits called ovarioles, whose number largely determines the number of eggs that can be potentially laid. Ovariole number in Drosophila is directly determined by the number of cellular structures called terminal filaments, which are stacks of cells that assemble in the larval ovary. Elucidating the developmental and regulatory mechanisms of terminal filament formation is thus key to understanding the regulation of insect reproduction through ovariole number regulation. We systematically measured mRNA expression of all cells in the larval ovary at the beginning, middle, and end of terminal filament formation. We also separated somatic and germ line cells during these stages and assessed their tissue-specific gene expression during larval ovary development. We found that the number of differentially expressed somatic genes is highest during the late stages of terminal filament formation and includes many signaling pathways that govern ovary development. We also show that germ line tissue, in contrast, shows greater differential expression during early stages of terminal filament formation, and highly expressed germ line genes at these stages largely control cell division and DNA repair. We provide a tissue-specific and temporal transcriptomic dataset of gene expression in the developing larval ovary as a resource to study insect reproduction.
Collapse
Affiliation(s)
- Shreeharsha Tarikere
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Guillem Ylla
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | - Cassandra G Extavour
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
18
|
Drosophila Keap1 xenobiotic response factor regulates developmental transcription through binding to chromatin. Dev Biol 2022; 481:139-147. [PMID: 34662537 PMCID: PMC9502878 DOI: 10.1016/j.ydbio.2021.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 01/03/2023]
Abstract
The Keap1-Nrf2 complex is a central regulator that mediates transcriptional responses to xenobiotic stimuli and is highly related with multiple human diseases. The molecular mechanisms and biological functions of Keap1 and Nrf2 are not fully understood. The Drosophila Keap1 homolog (dKeap1) is conserved with mammalian Keap1 except that dKeap1 contains a 156 aa C-terminal tail (CTD). A dKeap1 truncation with the CTD removed (dKeap1-ΔCTD) shows abolished nuclear localization and chromatin-binding. Expression of dKeap1-ΔCTD in the dKeap1 null background significantly rescues this mutant to the adult stage, but the files showed partial lethality, sterility and defects in adipose tissue. In the rescued flies, expression levels of ecdysone-response genes, ecdysone-synthetic genes and adipogenesis genes were down-regulated in specific tissues, indicating that the chromatin-binding of dKeap1 mediates the activation of these developmental genes. At the same time, dKeap1-ΔCTD can still suppress the basal expression of detoxifying genes and mediate the activation of these genes in response to xenobiotic stimuli, suggesting that the chromatin-binding of dKeap1 is not required for the regulation of detoxifying genes. These results support a model in which dKeap1 on one hand functions as an inhibitor for the Nrf2-mediated transcription in the xenobiotic response pathway and on the other hand functions as a chromatin-binding transcription activator in the developmental pathway. Our study reveals a novel mechanism whereby Keap1-Nrf2 xenobiotic response signaling regulates development using a mechanism independent of redox signaling.
Collapse
|
19
|
Hoshino R, Niwa R. Regulation of Mating-Induced Increase in Female Germline Stem Cells in the Fruit Fly Drosophila melanogaster. Front Physiol 2021; 12:785435. [PMID: 34950056 PMCID: PMC8689587 DOI: 10.3389/fphys.2021.785435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/17/2021] [Indexed: 01/19/2023] Open
Abstract
In many insect species, mating stimuli can lead to changes in various behavioral and physiological responses, including feeding, mating refusal, egg-laying behavior, energy demand, and organ remodeling, which are collectively known as the post-mating response. Recently, an increase in germline stem cells (GSCs) has been identified as a new post-mating response in both males and females of the fruit fly, Drosophila melanogaster. We have extensively studied mating-induced increase in female GSCs of D. melanogaster at the molecular, cellular, and systemic levels. After mating, the male seminal fluid peptide [e.g. sex peptide (SP)] is transferred to the female uterus. This is followed by binding to the sex peptide receptor (SPR), which evokes post-mating responses, including increase in number of female GSCs. Downstream of SP-SPR signaling, the following three hormones and neurotransmitters have been found to act on female GSC niche cells to regulate mating-induced increase in female GSCs: (1) neuropeptide F, a peptide hormone produced in enteroendocrine cells; (2) octopamine, a monoaminergic neurotransmitter synthesized in ovary-projecting neurons; and (3) ecdysone, a steroid hormone produced in ovarian follicular cells. These humoral factors are secreted from each organ and are received by ovarian somatic cells and regulate the strength of niche signaling in female GSCs. This review provides an overview of the latest findings on the inter-organ relationship to regulate mating-induced female GSC increase in D. melanogaster as a model. We also discuss the remaining issues that should be addressed in the future.
Collapse
Affiliation(s)
- Ryo Hoshino
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
20
|
Marca JEL, Somers WG. The Drosophila gonads: models for stem cell proliferation, self-renewal, and differentiation. AIMS GENETICS 2021. [DOI: 10.3934/genet.2014.1.55] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AbstractThe male and female gonads of Drosophila melanogaster have developed into powerful model systems for both the study of stem cell behaviours, and for understanding how stem cell misregulation can lead to cancers. Using these systems, one is able to observe and manipulate the resident stem cell populations in vivo with a great deal of licence. The tractability of the testis and ovary also allow researchers to explore a range of cellular mechanisms, such as proliferation and polarity, as well as the influence exerted by the local environment through a host of highly-conserved signalling pathways. Importantly, many of the cellular behaviours and processes studied in the Drosophila testis and ovary are known to be disrupted, or otherwise misregulated, in human tumourigenic cells. Here, we review the mechanisms relating to stem cell behaviour, though we acknowledge there are many other fascinating aspects of gametogenesis, including the invasive behaviour of migratory border cells in the Drosophila ovary that, though relevant to the study of tumourigenesis, will unfortunately not be covered.
Collapse
Affiliation(s)
- John E. La Marca
- Department of Genetics, La Trobe University, Melbourne, VIC 3086, Australia
| | | |
Collapse
|
21
|
Hu X, Li M, Hao X, Lu Y, Zhang L, Wu G. The Osa-Containing SWI/SNF Chromatin-Remodeling Complex Is Required in the Germline Differentiation Niche for Germline Stem Cell Progeny Differentiation. Genes (Basel) 2021; 12:genes12030363. [PMID: 33806269 PMCID: PMC7998989 DOI: 10.3390/genes12030363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/22/2022] Open
Abstract
The Drosophila ovary is recognized as a powerful model to study stem cell self-renewal and differentiation. Decapentaplegic (Dpp) is secreted from the germline stem cell (GSC) niche to activate Bone Morphogenic Protein (BMP) signaling in GSCs for their self-renewal and is restricted in the differentiation niche for daughter cell differentiation. Here, we report that Switch/sucrose non-fermentable (SWI/SNF) component Osa depletion in escort cells (ECs) results in a blockage of GSC progeny differentiation. Further molecular and genetic analyses suggest that the defective germline differentiation is partially attributed to the elevated dpp transcription in ECs. Moreover, ectopic Engrailed (En) expression in osa-depleted ECs partially contributes to upregulated dpp transcription. Furthermore, we show that Osa regulates germline differentiation in a Brahma (Brm)-associated protein (BAP)-complex-dependent manner. Additionally, the loss of EC long cellular processes upon osa depletion may also partly contribute to the germline differentiation defect. Taken together, these data suggest that the epigenetic factor Osa plays an important role in controlling EC characteristics and germline lineage differentiation.
Collapse
Affiliation(s)
- Xiaolong Hu
- State Key Laboratory of Microbial Metabolism, The Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; (X.H.); (M.L.)
| | - Mengjie Li
- State Key Laboratory of Microbial Metabolism, The Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; (X.H.); (M.L.)
| | - Xue Hao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; (X.H.); (Y.L.); (L.Z.)
| | - Yi Lu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; (X.H.); (Y.L.); (L.Z.)
| | - Lei Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China; (X.H.); (Y.L.); (L.Z.)
- School of Life Science and Technology, Shanghai Tech University, Shanghai 201210, China
- Bio-Research Innovation Center, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Suzhou 215121, China
| | - Geng Wu
- State Key Laboratory of Microbial Metabolism, The Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences &Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; (X.H.); (M.L.)
- Correspondence:
| |
Collapse
|
22
|
A Progressive Somatic Cell Niche Regulates Germline Cyst Differentiation in the Drosophila Ovary. Curr Biol 2021; 31:840-852.e5. [PMID: 33340458 DOI: 10.1016/j.cub.2020.11.053] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 10/02/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022]
Abstract
In the germarium of the Drosophila ovary, developing germline cysts are surrounded by a population of somatic escort cells that are known to function as the niche cells for germline differentiation;1 however, the underlying molecular mechanisms of this niche function remain poorly understood. Through single-cell gene expression profiling combined with genetic analyses, we here demonstrate that the escort cells can be spatially and functionally divided into two successive domains. The anterior escort cells (aECs) specifically produce ecdysone, which acts on the cystoblast to promote synchronous cell division, whereas the posterior escort cells (pECs) respond to ecdysone signaling and regulate soma-germline cell adhesion to promote the transition from 16-cell cyst-to-egg chamber formation. The patterning of the aEC and pEC domains is independent of the germline but is dependent on JAK/STAT signaling activity, which emanates from the posterior. Thus, a heterogeneous population of escort cells constitutes a stepwise niche environment to orchestrate cystoblast division and differentiation toward egg chamber formation.
Collapse
|
23
|
Finger DS, Whitehead KM, Phipps DN, Ables ET. Nuclear receptors linking physiology and germline stem cells in Drosophila. VITAMINS AND HORMONES 2021; 116:327-362. [PMID: 33752824 PMCID: PMC8063499 DOI: 10.1016/bs.vh.2020.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Maternal nutrition and physiology are intimately associated with reproductive success in diverse organisms. Despite decades of study, the molecular mechanisms linking maternal diet to the production and quality of oocytes remain poorly defined. Nuclear receptors (NRs) link nutritional signals to cellular responses and are essential for oocyte development. The fruit fly, Drosophila melanogaster, is an excellent genetically tractable model to study the relationship between NR signaling and oocyte production. In this review, we explore how NRs in Drosophila regulate the earliest stages of oocyte development. Long-recognized as an essential mediator of developmental transitions, we focus on the intrinsic roles of the Ecdysone Receptor and its ligand, ecdysone, in oogenesis. We also review recent studies suggesting broader roles for NRs as regulators of maternal physiology and their impact specifically on oocyte production. We propose that NRs form the molecular basis of a broad physiological surveillance network linking maternal diet with oocyte production. Given the functional conservation between Drosophila and humans, continued experimental investigation into the molecular mechanisms by which NRs promote oogenesis will likely aid our understanding of human fertility.
Collapse
Affiliation(s)
- Danielle S Finger
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Kaitlin M Whitehead
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Daniel N Phipps
- Department of Biology, East Carolina University, Greenville, NC, United States
| | - Elizabeth T Ables
- Department of Biology, East Carolina University, Greenville, NC, United States.
| |
Collapse
|
24
|
Tu R, Duan B, Song X, Chen S, Scott A, Hall K, Blanck J, DeGraffenreid D, Li H, Perera A, Haug J, Xie T. Multiple Niche Compartments Orchestrate Stepwise Germline Stem Cell Progeny Differentiation. Curr Biol 2020; 31:827-839.e3. [PMID: 33357404 DOI: 10.1016/j.cub.2020.12.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 11/17/2020] [Accepted: 12/15/2020] [Indexed: 11/28/2022]
Abstract
The niche controls stem cell self-renewal and progenitor differentiation for maintaining adult tissue homeostasis in various organisms. However, it remains unclear whether the niche is compartmentalized to control stem cell self-renewal and stepwise progeny differentiation. In the Drosophila ovary, inner germarial sheath (IGS) cells form a niche for controlling germline stem cell (GSC) progeny differentiation. In this study, we have identified four IGS subpopulations, which form linearly arranged niche compartments for controlling GSC maintenance and multi-step progeny differentiation. Single-cell analysis of the adult ovary has identified four IGS subpopulations (IGS1-IGS4), the identities and cellular locations of which have been further confirmed by fluorescent in situ hybridization. IGS1 and IGS2 physically interact with GSCs and mitotic cysts to control GSC maintenance and cyst formation, respectively, whereas IGS3 and IGS4 physically interact with 16-cell cysts to regulate meiosis, oocyte development, and cyst morphological change. Finally, one follicle cell progenitor population has also been transcriptionally defined for facilitating future studies on follicle stem cell regulation. Therefore, this study has structurally revealed that the niche is organized into multiple compartments for orchestrating stepwise adult stem cell development and has also provided useful resources and tools for further functional characterization of the niche in the future.
Collapse
Affiliation(s)
- Renjun Tu
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Bo Duan
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Xiaoqing Song
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Shiyuan Chen
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Allison Scott
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Kate Hall
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Jillian Blanck
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Dustin DeGraffenreid
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Hua Li
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Anoja Perera
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Jeff Haug
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA
| | - Ting Xie
- Stowers Institute for Medical Research, 1000 East 50th Street, Kansas City, MO 64110, USA; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
25
|
Weaver LN, Drummond-Barbosa D. The Nuclear Receptor Seven Up Regulates Genes Involved in Immunity and Xenobiotic Response in the Adult Drosophila Female Fat Body. G3 (BETHESDA, MD.) 2020; 10:4625-4635. [PMID: 33087412 PMCID: PMC7718730 DOI: 10.1534/g3.120.401745] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/17/2020] [Indexed: 01/02/2023]
Abstract
The physiology of organisms depends on inter-organ communication in response to changes in the environment. Nuclear receptors are broadly expressed transcription factors that respond to circulating molecules to control many biological processes, including immunity, detoxification, and reproduction. Although the tissue-intrinsic roles of nuclear receptors in reproduction have been extensively studied, there is increasing evidence that nuclear receptor signaling in peripheral tissues can also influence oogenesis. We previously showed that the Drosophila nuclear receptor Seven up (Svp) is required in the adult fat body to regulate distinct steps of oogenesis; however, the relevant downstream targets of Svp remain unknown. Here, we took an RNA sequencing approach to identify candidate Svp targets specifically in the adult female fat body that might mediate this response. svp knockdown in the adult female fat body significantly downregulated immune genes involved in the first line of pathogen defense, suggesting a role for Svp in stimulating early immunity. In addition, we found that Svp transcriptionally regulates genes involved in each step of the xenobiotic detoxification response. Based on these findings, we propose a testable model in which Svp functions in the adult female fat body to stimulate early defense against pathogens and facilitate detoxification as part of its mechanisms to promote oogenesis.
Collapse
Affiliation(s)
- Lesley N Weaver
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
| | - Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
| |
Collapse
|
26
|
Yoshinari Y, Ameku T, Kondo S, Tanimoto H, Kuraishi T, Shimada-Niwa Y, Niwa R. Neuronal octopamine signaling regulates mating-induced germline stem cell increase in female Drosophila melanogaster. eLife 2020; 9:57101. [PMID: 33077027 PMCID: PMC7591258 DOI: 10.7554/elife.57101] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022] Open
Abstract
Stem cells fuel the development and maintenance of tissues. Many studies have addressed how local signals from neighboring niche cells regulate stem cell identity and their proliferative potential. However, the regulation of stem cells by tissue-extrinsic signals in response to environmental cues remains poorly understood. Here we report that efferent octopaminergic neurons projecting to the ovary are essential for germline stem cell (GSC) increase in response to mating in female Drosophila. The neuronal activity of the octopaminergic neurons is required for mating-induced GSC increase as they relay the mating signal from sex peptide receptor-positive cholinergic neurons. Octopamine and its receptor Oamb are also required for mating-induced GSC increase via intracellular Ca2+ signaling. Moreover, we identified Matrix metalloproteinase-2 as a downstream component of the octopamine-Ca2+ signaling to induce GSC increase. Our study provides a mechanism describing how neuronal system couples stem cell behavior to environmental cues through stem cell niche signaling.
Collapse
Affiliation(s)
- Yuto Yoshinari
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tomotsune Ameku
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Shu Kondo
- Invertebrate Genetics Laboratory, National Institute of Genetics, Mishima, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Takayuki Kuraishi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.,AMED-PRIME, Japan Agency for Medical Research and Development, Tokyo, Japan
| | - Yuko Shimada-Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
27
|
Zipper L, Jassmann D, Burgmer S, Görlich B, Reiff T. Ecdysone steroid hormone remote controls intestinal stem cell fate decisions via the PPARγ-homolog Eip75B in Drosophila. eLife 2020; 9:e55795. [PMID: 32773037 PMCID: PMC7440922 DOI: 10.7554/elife.55795] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 08/07/2020] [Indexed: 12/27/2022] Open
Abstract
Developmental studies revealed fundamental principles on how organ size and function is achieved, but less is known about organ adaptation to new physiological demands. In fruit flies, juvenile hormone (JH) induces intestinal stem cell (ISC) driven absorptive epithelial expansion balancing energy uptake with increased energy demands of pregnancy. Here, we show 20-Hydroxy-Ecdysone (20HE)-signaling controlling organ homeostasis with physiological and pathological implications. Upon mating, 20HE titer in ovaries and hemolymph are increased and act on nearby midgut progenitors inducing Ecdysone-induced-protein-75B (Eip75B). Strikingly, the PPARγ-homologue Eip75B drives ISC daughter cells towards absorptive enterocyte lineage ensuring epithelial growth. To our knowledge, this is the first time a systemic hormone is shown to direct local stem cell fate decisions. Given the protective, but mechanistically unclear role of steroid hormones in female colorectal cancer patients, our findings suggest a tumor-suppressive role for steroidal signaling by promoting postmitotic fate when local signaling is deteriorated.
Collapse
Affiliation(s)
- Lisa Zipper
- Institute of Genetics, Heinrich-Heine-UniversityDüsseldorfGermany
| | - Denise Jassmann
- Institute of Genetics, Heinrich-Heine-UniversityDüsseldorfGermany
| | - Sofie Burgmer
- Institute of Genetics, Heinrich-Heine-UniversityDüsseldorfGermany
| | - Bastian Görlich
- Institute of Genetics, Heinrich-Heine-UniversityDüsseldorfGermany
| | - Tobias Reiff
- Institute of Genetics, Heinrich-Heine-UniversityDüsseldorfGermany
| |
Collapse
|
28
|
Ramos S, Chelemen F, Pagone V, Elshaer N, Irles P, Piulachs MD. Eyes absent in the cockroach panoistic ovaries regulates proliferation and differentiation through ecdysone signalling. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 123:103407. [PMID: 32417417 DOI: 10.1016/j.ibmb.2020.103407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 04/26/2020] [Accepted: 05/10/2020] [Indexed: 06/11/2023]
Abstract
Eyes absent (Eya), is a protein structurally conserved from hydrozoans to humans, for which two basic roles have been reported: it can act as a transcription cofactor and as a protein tyrosine phosphatase. Eya was discovered in the fly Drosophila melanogaster in relation to its function in eye development, and the same function was later reported in other insects. Eya is also involved in insect oogenesis, although studies in this sense are limited to D. melanogaster, which has meroistic ovaries, and where eya mutations abolish gonad formation. In the present work we studied the function of eya in the panoistic ovary of the cockroach Blattella germanica. We show that eya is essential for correct development of panoistic ovaries. In B. germanica, eya acts at different level and in a distinct way in the germarium and the vitellarium. In the germarium, eya contributes to maintain the correct number of somatic and germinal cells by regulating the expression of steroidogenic genes in the ovary. In the vitellarium, eya facilitates follicle cells proliferation and contributes to regulate the cell program, in the context of basal ovarian follicle maturation. Thus, eya-depleted females of B. germanica arrest the growth and maturation of basal ovarian follicles and become sterile.
Collapse
Affiliation(s)
- S Ramos
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain
| | - F Chelemen
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain
| | - V Pagone
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain
| | - N Elshaer
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain; Department of Plant Protection, Faculty of Agriculture, Zagazig University, Egypt
| | - P Irles
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain; Instituto de Ciencias Agronomicas y Veterinarias, Universidad de O'Higgins, Chile
| | - M D Piulachs
- Institut de Biologia Evolutiva (CSIC- Universitat Pompeu Fabra), Passeig Maritim de la Barceloneta, 37, 08003, Barcelona, Spain.
| |
Collapse
|
29
|
Yatsenko AS, Kucherenko MM, Xie Y, Aweida D, Urlaub H, Scheibe RJ, Cohen S, Shcherbata HR. Profiling of the muscle-specific dystroglycan interactome reveals the role of Hippo signaling in muscular dystrophy and age-dependent muscle atrophy. BMC Med 2020; 18:8. [PMID: 31959160 PMCID: PMC6971923 DOI: 10.1186/s12916-019-1478-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Dystroglycanopathies are a group of inherited disorders characterized by vast clinical and genetic heterogeneity and caused by abnormal functioning of the ECM receptor dystroglycan (Dg). Remarkably, among many cases of diagnosed dystroglycanopathies, only a small fraction can be linked directly to mutations in Dg or its regulatory enzymes, implying the involvement of other, not-yet-characterized, Dg-regulating factors. To advance disease diagnostics and develop new treatment strategies, new approaches to find dystroglycanopathy-related factors should be considered. The Dg complex is highly evolutionarily conserved; therefore, model genetic organisms provide excellent systems to address this challenge. In particular, Drosophila is amenable to experiments not feasible in any other system, allowing original insights about the functional interactors of the Dg complex. METHODS To identify new players contributing to dystroglycanopathies, we used Drosophila as a genetic muscular dystrophy model. Using mass spectrometry, we searched for muscle-specific Dg interactors. Next, in silico analyses allowed us to determine their association with diseases and pathological conditions in humans. Using immunohistochemical, biochemical, and genetic interaction approaches followed by the detailed analysis of the muscle tissue architecture, we verified Dg interaction with some of the discovered factors. Analyses of mouse muscles and myocytes were used to test if interactions are conserved in vertebrates. RESULTS The muscle-specific Dg complexome revealed novel components that influence the efficiency of Dg function in the muscles. We identified the closest human homologs for Dg-interacting partners, determined their significant enrichment in disease-associations, and verified some of the newly identified Dg interactions. We found that Dg associates with two components of the mechanosignaling Hippo pathway: the WW domain-containing proteins Kibra and Yorkie. Importantly, this conserved interaction manages adult muscle size and integrity. CONCLUSIONS The results presented in this study provide a new list of muscle-specific Dg interactors, further analysis of which could aid not only in the diagnosis of muscular dystrophies, but also in the development of new therapeutics. To regulate muscle fitness during aging and disease, Dg associates with Kibra and Yorkie and acts as a transmembrane Hippo signaling receptor that transmits extracellular information to intracellular signaling cascades, regulating muscle gene expression.
Collapse
Affiliation(s)
- Andriy S Yatsenko
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | - Mariya M Kucherenko
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: Department of Cardiothoracic and Vascular Surgery, German Heart Center Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Institute of Physiology, Charité - University Medicine Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Yuanbin Xie
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Present Address: University Medical Center, Centre for Anatomy, Institute of Neuroanatomy, Georg-August-University Göttingen, Kreuzbergring 36, 37075, Göttingen, Germany
| | - Dina Aweida
- Faculty of Biology, Technion, 32000, Haifa, Israel
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Research Group, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.,Bioanalytics Institute for Clinical Chemistry, University Medical Center Goettingen, Robert Koch Strasse 40, 37075, Göttingen, Germany
| | - Renate J Scheibe
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany
| | | | - Halyna R Shcherbata
- Gene Expression and Signaling Group, Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625, Hannover, Germany. .,Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
30
|
Weaver LN, Drummond-Barbosa D. The nuclear receptor seven up functions in adipocytes and oenocytes to control distinct steps of Drosophila oogenesis. Dev Biol 2019; 456:179-189. [PMID: 31470019 PMCID: PMC6884690 DOI: 10.1016/j.ydbio.2019.08.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 08/23/2019] [Indexed: 02/09/2023]
Abstract
Reproduction is intimately linked to the physiology of an organism. Nuclear receptors are widely expressed transcription factors that mediate the effects of many circulating molecules on physiology and reproduction. While multiple studies have focused on the roles of nuclear receptors intrinsically in the ovary, it remains largely unknown how the actions of nuclear receptors in peripheral tissues influence oogenesis. We identified the nuclear receptor encoded by svp as a novel regulator of oogenesis in adult Drosophila. Global somatic knockdown of svp reduces egg production by increasing GSC loss, death of early germline cysts, and degeneration of vitellogenic follicles. Tissue-specific knockdown experiments revealed that svp remotely controls these different steps of oogenesis through separate mechanisms involving distinct tissues. Specifically, adipocyte-specific svp knockdown impairs GSC maintenance and early germline cyst survival, whereas oenocyte-specific svp knockdown increases the death of vitellogenic follicles without any effects on GSCs or early cysts. These results illustrate that nuclear receptors can control reproduction through a variety of mechanisms involving peripheral tissues.
Collapse
Affiliation(s)
- Lesley N Weaver
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, 21205, USA.
| |
Collapse
|
31
|
McDonald SI, Beachum AN, Hinnant TD, Blake AJ, Bynum T, Hickman EP, Barnes J, Churchill KL, Roberts TS, Zangwill DE, Ables ET. Novel cis-regulatory regions in ecdysone responsive genes are sufficient to promote gene expression in Drosophila ovarian cells. Gene Expr Patterns 2019; 34:119074. [PMID: 31563631 PMCID: PMC6996244 DOI: 10.1016/j.gep.2019.119074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 06/18/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022]
Abstract
The insect steroid hormone ecdysone is a key regulator of oogenesis in Drosophila melanogaster and many other species. Despite the diversity of cellular functions of ecdysone in oogenesis, the molecular regulation of most ecdysone-responsive genes in ovarian cells remains largely unexplored. We performed a functional screen using the UAS/Gal4 system to identify non-coding cis-regulatory elements within well-characterized ecdysone-response genes capable of driving transcription of an indelible reporter in ovarian cells. Using two publicly available transgenic collections (the FlyLight and Vienna Tiles resources), we tested 62 Gal4 drivers corresponding to ecdysone-response genes EcR, usp, E75, br, ftz-f1 and Hr3. We observed 31 lines that were sufficient to drive a UAS-lacZ reporter in discrete cell populations in the ovary. Reporter expression was reproducibly observed in both somatic and germ cells at distinct stages of oogenesis, including those previously characterized as critical points of ecdysone regulation. Our studies identified several useful new reagents, adding to the UAS/Gal4 toolkit available for genetic analysis of oogenesis in Drosophila. Further, our study provides novel insight into the molecular regulation of ecdysone signaling in oogenesis.
Collapse
Affiliation(s)
| | - Allison N Beachum
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Taylor D Hinnant
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Amelia J Blake
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Tierra Bynum
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - E Parris Hickman
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Joseph Barnes
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Kaely L Churchill
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Tamesia S Roberts
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Denise E Zangwill
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Elizabeth T Ables
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA.
| |
Collapse
|
32
|
Hsu HJ, Bahader M, Lai CM. Molecular control of the female germline stem cell niche size in Drosophila. Cell Mol Life Sci 2019; 76:4309-4317. [PMID: 31300869 PMCID: PMC11105562 DOI: 10.1007/s00018-019-03223-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 06/17/2019] [Accepted: 07/05/2019] [Indexed: 11/26/2022]
Abstract
Adult stem cells have a unique capacity to renew themselves and generate differentiated cells that are needed in the body. These cells are recruited and maintained by the surrounding microenvironment, known as the stem cell niche, during organ development. Thus, the stem cell niche is required for proper tissue homeostasis, and its dysregulation is associated with tumorigenesis and tissue degeneration. The identification of niche components and the mechanisms that regulate niche establishment and maintenance, however, are just beginning to be uncovered. Germline stem cells (GSCs) of the Drosophila ovary provide an excellent model for studying the stem cell niche in vivo because of their well-characterized cell biology and the availability of genetic tools. In this review, we introduce the ovarian GSC niche, and the key signaling pathways for niche precursor segregation, niche specification, and niche extracellular environment establishment and niche maintenance that are involved in regulating niche size during development and adulthood.
Collapse
Affiliation(s)
- Hwei-Jan Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan.
| | - Majid Bahader
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
| | - Chun-Ming Lai
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, 11529, Taiwan
- Department of Developmental Biology, Sloan-Kettering Institute, 1275 York Ave, New York, NY, 10065, USA
| |
Collapse
|
33
|
Xu QY, Deng P, Mu LL, Fu KY, Guo WC, Li GQ. Silencing Taiman impairs larval development in Leptinotarsa decemlineata. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2019; 160:30-39. [PMID: 31519255 DOI: 10.1016/j.pestbp.2019.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 06/19/2019] [Accepted: 06/23/2019] [Indexed: 06/10/2023]
Abstract
An exploration of novel control strategies for Leptinotarsa decemlineata is becoming more pressing given rapid evolution of insecticide resistance and rise of production loss of potato. Dietary delivery of bacterially expressed double-stranded RNA (dsRNA) is a promising alternative for management. An important first step is to uncover possible RNA-interference (RNAi)-target genes effective against both young and old larvae. Taiman (Tai) is a basic-helix-loop-helix/Per-Arnt-Sim transcription factor that is involved in the mediation of both juvenile hormone (JH) and 20-hydroxyecdysone (20E) signaling. In the present paper, we found that continuous ingestion of dsTai for three days by third (penultimate)-instar larvae caused approximately 20% larval mortality and 80% pupation failure. The larval lethality resulted from failed cuticle and tracheae shedding, which subsequently reduced foliage consumption and nutrient absorption, and depleted lipid stores. In contrast, pupation failure derived from disturbed JH and 20E signals, and disordered nutrient homeostasis including, among others, inhibition of trehalose metabolism and reduction of chitin content. Knockdown of LdTai caused similar larval lethality and pupation impairment in second and fourth (final) larval instars. Therefore, LdTai is among the most attractive candidate genes for RNAi to control L. decemlineata larvae.
Collapse
Affiliation(s)
- Qing-Yu Xu
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Pan Deng
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China
| | - Li-Li Mu
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| | - Kai-Yun Fu
- Institute of Plant Protection, Xinjiang Academy of Agricultural Sciences, Urumqi 830091, China; Key Laboratory of Intergraded Management of Harmful Crop Vermin of China North-western Oasis, Ministry of Agriculture, China
| | - Wen-Chao Guo
- Institute of Microbiological Application, Xinjiang, Academy of Agricultural Science; Urumqi, 830091, China
| | - Guo-Qing Li
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
34
|
Drummond-Barbosa D. Local and Physiological Control of Germline Stem Cell Lineages in Drosophila melanogaster. Genetics 2019; 213:9-26. [PMID: 31488592 PMCID: PMC6727809 DOI: 10.1534/genetics.119.300234] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022] Open
Abstract
The long-term survival of any multicellular species depends on the success of its germline in producing high-quality gametes and maximizing survival of the offspring. Studies in Drosophila melanogaster have led our growing understanding of how germline stem cell (GSC) lineages maintain their function and adjust their behavior according to varying environmental and/or physiological conditions. This review compares and contrasts the local regulation of GSCs by their specialized microenvironments, or niches; discusses how diet and diet-dependent factors, mating, and microorganisms modulate GSCs and their developing progeny; and briefly describes the tie between physiology and development during the larval phase of the germline cycle. Finally, it concludes with broad comparisons with other organisms and some future directions for further investigation.
Collapse
Affiliation(s)
- Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland 21205
| |
Collapse
|
35
|
Li M, Hu X, Zhang S, Ho MS, Wu G, Zhang L. Traffic jam regulates the function of the ovarian germline stem cell progeny differentiation niche during pre-adult stage in Drosophila. Sci Rep 2019; 9:10124. [PMID: 31300663 PMCID: PMC6626045 DOI: 10.1038/s41598-019-45317-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 03/29/2019] [Indexed: 11/28/2022] Open
Abstract
Stem cell self-renewal and the daughter cell differentiation are tightly regulated by the respective niches, which produce extrinsic cues to support the proper development. In Drosophila ovary, Dpp is secreted from germline stem cell (GSC) niche and activates the BMP signaling in GSCs for their self-renewal. Escort cells (ECs) in differentiation niche restrict Dpp outside the GSC niche and extend protrusions to help with proper differentiation of the GSC daughter cells. Here we provide evidence that loss of large Maf transcriptional factor Traffic jam (Tj) blocks GSC progeny differentiation. Spatio-temporal specific knockdown experiments indicate that Tj is required in pre-adult EC lineage for germline differentiation control. Further molecular and genetic analyses suggest that the defective germline differentiation caused by tj-depletion is partly attributed to the elevated dpp in the differentiation niche. Moreover, our study reveals that tj-depletion induces ectopic En expression outside the GSC niche, which contributes to the upregulated dpp expression in ECs as well as GSC progeny differentiation defect. Alternatively, loss of EC protrusions and decreased EC number elicited by tj-depletion may also partially contribute to the germline differentiation defect. Collectively, our findings suggest that Tj in ECs regulates germline differentiation by controlling the differentiation niche characteristics.
Collapse
Affiliation(s)
- Mengjie Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, The Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaolong Hu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, The Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shu Zhang
- School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Margaret S Ho
- School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Geng Wu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences & Biotechnology, The Joint International Research Laboratory of Metabolic & Developmental Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Lei Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China. .,School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China.
| |
Collapse
|
36
|
Xu QY, Du JL, Mu LL, Guo WC, Li GQ. Importance of Taiman in Larval-Pupal Transition in Leptinotarsa decemlineata. Front Physiol 2019; 10:724. [PMID: 31263425 PMCID: PMC6584964 DOI: 10.3389/fphys.2019.00724] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 05/27/2019] [Indexed: 02/01/2023] Open
Abstract
Insect Taiman (Tai) binds to methoprene-tolerant to form a heterodimeric complex, mediating juvenile hormone (JH) signaling to regulate larval development and to prevent premature metamorphosis. Tai also acts as a steroid receptor coactivator of 20-hydroxyecdysone (20E) receptor heterodimer, ecdysone receptor (EcR) and Ultraspiracle (USP), to control the differentiation of early germline cells and the migration of specific follicle cells and border cells in ovaries in several insect species. In holometabolous insects, however, whether Tai functions as the coactivator of EcR/USP to transduce 20E message during larval-pupal transition is unknown. In the present paper, we found that the LdTai mRNA levels were positively correlated with circulating JH and 20E titers in Leptinotarsa decemlineata; and ingestion of either JH or 20E stimulated the transcription of LdTai. Moreover, RNA interference (RNAi)-aided knockdown of LdTai at the fourth (final) instar stage repressed both JH and 20E signals, inhibited larval growth and shortened larval developing period. The knockdown caused 100% larval lethality due to failure of larval-pupal ecdysis. Under the apolysed larval cuticle, the LdTai RNAi prepupae possessed pupal thorax. In contrast, the process of tracheal ecdysis was uncompleted. Neither JH nor 20E rescued the aforementioned defectives in LdTai RNAi larvae. It appears that Tai mediates both JH and 20E signaling. Our results uncover a link between JH and 20E pathways during metamorphosis in L. decemlineata.
Collapse
Affiliation(s)
- Qing-Yu Xu
- Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Jun-Li Du
- Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, College of Plant Protection, Nanjing Agricultural University, Nanjing, China.,College of Agriculture, Anhui Science and Technology University, Fengyang, China
| | - Li-Li Mu
- Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Wen-Chao Guo
- Institute of Plant Protection, Xinjiang Academy of Agricultural Sciences, Urumqi, China.,Key Laboratory of Intergraded Management of Harmful Crop Vermin of China North-Western Oasis, Ministry of Agriculture, Urumqi, China
| | - Guo-Qing Li
- Key Laboratory of Integrated Management of Crop Diseases and Pests, Ministry of Education, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
37
|
Evolution of sexually-transferred steroids and mating-induced phenotypes in Anopheles mosquitoes. Sci Rep 2019; 9:4669. [PMID: 30874601 PMCID: PMC6420574 DOI: 10.1038/s41598-019-41094-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/28/2019] [Indexed: 11/08/2022] Open
Abstract
Human malaria, which remains a major public health problem, is transmitted by a subset of Anopheles mosquitoes belonging to only three out of eight subgenera: Anopheles, Cellia and Nyssorhynchus. Unlike almost every other insect species, males of some Anopheles species produce steroid hormones which are transferred to females during copulation to influence their reproduction. Steroids are consequently a potential target for malaria vector control. Here, we analysed the evolution of sexually-transferred steroids and their effects on female reproductive traits across Anopheles by using a set of 16 mosquito species (five Anopheles, eight Cellia, and three Nyssorhynchus), including malaria vector and non-vector species. We show that male steroid production and transfer are specific to the Cellia and therefore represent a synapomorphy of this subgenus. Furthermore, we show that mating-induced effects in females are variable across species and differences are not correlated with sexually-transferred steroids or with Anopheles ability to transmit human malaria. Overall, our findings highlight that Anopheles mosquitoes have evolved different reproductive strategies, independently of being a malaria vector or not.
Collapse
|
38
|
Swevers L. An update on ecdysone signaling during insect oogenesis. CURRENT OPINION IN INSECT SCIENCE 2019; 31:8-13. [PMID: 31109678 DOI: 10.1016/j.cois.2018.07.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/04/2018] [Indexed: 06/09/2023]
Abstract
An overview is presented of the different functions of ecdysone signaling during insect oogenesis. An extensive genetic toolkit allowed analysis with unprecedented temporal and spatial detail in Drosophila where functions were revealed in stem cell proliferation and niche maintenance, germline cyst differentiation and follicle formation, integration of nutrient and lipid signaling, follicle maturation and ovulation. Besides putative autocrine/paracrine signaling, hormonal networks were identified that integrate ecdysone with other endocrine signaling pathways. In other insects, progress in oogenesis has lagged behind although recently RNAi emerged as a new tool to analyze gene function in ovaries in hemimetabolous insects and Tribolium.
Collapse
Affiliation(s)
- Luc Swevers
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences & Applications, NCSR "Demokritos", Aghia Paraskevi, Greece.
| |
Collapse
|
39
|
Yoshinari Y, Kurogi Y, Ameku T, Niwa R. Endocrine regulation of female germline stem cells in the fruit fly Drosophila melanogaster. CURRENT OPINION IN INSECT SCIENCE 2019; 31:14-19. [PMID: 31109668 DOI: 10.1016/j.cois.2018.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/27/2018] [Accepted: 07/03/2018] [Indexed: 06/09/2023]
Abstract
Germline stem cells (GSCs) are critical for the generation of sperms and eggs in most animals including the fruit fly Drosophila melanogaster. It is well known that self-renewal and differentiation of female D. melanogaster GSCs are regulated by local niche signals. However, little is known about whether and how the GSC number is regulated by paracrine signals. In the last decade, however, multiple humoral factors, including insulin and ecdysteroids, have been recognized as key regulators of female D. melanogaster GSCs. This review paper summarizes the role of humoral factors in female D. melanogaster GSC proliferation and maintenance in response to internal and external conditions, such as nutrients, mating stimuli, and aging.
Collapse
Affiliation(s)
- Yuto Yoshinari
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8572, Japan
| | - Yoshitomo Kurogi
- College of Biological Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8572, Japan
| | - Tomotsune Ameku
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8572, Japan
| | - Ryusuke Niwa
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8572, Japan; AMED-CREST, Japan Agency for Medical Research and Development, 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan.
| |
Collapse
|
40
|
Shcherbata HR. miRNA functions in stem cells and their niches: lessons from the Drosophila ovary. CURRENT OPINION IN INSECT SCIENCE 2019; 31:29-36. [PMID: 31109670 DOI: 10.1016/j.cois.2018.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 06/09/2023]
Abstract
From the very beginning of the miRNA era, Drosophila has served as an excellent model for explanation of miRNA biogenesis. Now Drosophila continues to be used in numerous studies aiming to decipher biological roles of individual miRNAs in a living organism. MiRNAs have emerged as an important regulatory class that adjusts gene expression in response to stress; therefore, it is particularly important to elucidate miRNA-based regulatory networks that appear in response to fluctuations in intrinsic and extrinsic environments. This review explores the major advances in understanding condition-dependent roles of miRNAs in adult stem cell biology using the Drosophila ovarian germline stem cell niche community as a model system.
Collapse
Affiliation(s)
- Halyna R Shcherbata
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany; Institute of Cell Biochemistry, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany.
| |
Collapse
|
41
|
Mirth CK, Nogueira Alves A, Piper MD. Turning food into eggs: insights from nutritional biology and developmental physiology of Drosophila. CURRENT OPINION IN INSECT SCIENCE 2019; 31:49-57. [PMID: 31109673 DOI: 10.1016/j.cois.2018.08.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/27/2018] [Indexed: 06/09/2023]
Abstract
Nutrition plays a central role in fecundity, regulating the onset of reproductive maturity, egg production, and the survival and health of offspring from insects to humans. Although decades of research have worked to uncover how nutrition mediates these effects, it has proven difficult to disentangle the relative role of nutrients as the raw material for egg and offspring development versus their role in stimulating endocrine cascades necessary to drive development. This has been further complicated by the fact that both nutrients and the signalling cascades they regulate interact in complex ways to control fecundity. Separating the two effects becomes important when trying to understand how fecundity is regulated, and in devising strategies to offset the negative effects of nutrition on reproductive health. In this review, we use the extensive literature on egg development in the fruit fly Drosophila melanogaster to explore how the nutrients from food provide the building blocks and stimulate signalling cascades necessary for making an egg.
Collapse
Affiliation(s)
- Christen K Mirth
- School of Biological Sciences, Monash University, Melbourne, Victoria 3800, Australia.
| | - André Nogueira Alves
- School of Biological Sciences, Monash University, Melbourne, Victoria 3800, Australia
| | - Matthew Dw Piper
- School of Biological Sciences, Monash University, Melbourne, Victoria 3800, Australia
| |
Collapse
|
42
|
Nganga R, Oleinik N, Kim J, Selvam SP, De Palma R, Johnson KA, Parikh RY, Gangaraju V, Peterson Y, Dany M, Stahelin RV, Voelkel-Johnson C, Szulc ZM, Bieberich E, Ogretmen B. Receptor-interacting Ser/Thr kinase 1 (RIPK1) and myosin IIA-dependent ceramidosomes form membrane pores that mediate blebbing and necroptosis. J Biol Chem 2018; 294:502-519. [PMID: 30420430 DOI: 10.1074/jbc.ra118.005865] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/08/2018] [Indexed: 12/29/2022] Open
Abstract
Formation of membrane pores/channels regulates various cellular processes, such as necroptosis or stem cell niche signaling. However, the roles of membrane lipids in the formation of pores and their biological functions are largely unknown. Here, using the cellular stress model evoked by the sphingolipid analog drug FTY720, we show that formation of ceramide-enriched membrane pores, referred to here as ceramidosomes, is initiated by a receptor-interacting Ser/Thr kinase 1 (RIPK1)-ceramide complex transported to the plasma membrane by nonmuscle myosin IIA-dependent trafficking in human lung cancer cells. Molecular modeling/simulation coupled with site-directed mutagenesis revealed that Asp147 or Asn169 of RIPK1 are key for ceramide binding and that Arg258 or Leu293 residues are involved in the myosin IIA interaction, leading to ceramidosome formation and necroptosis. Moreover, generation of ceramidosomes independently of any external drug/stress stimuli was also detected in the plasma membrane of germ line stem cells in ovaries during the early stages of oogenesis in Drosophila melanogaster Inhibition of ceramidosome formation via myosin IIA silencing limited germ line stem cell signaling and abrogated oogenesis. In conclusion, our findings indicate that the RIPK1-ceramide complex forms large membrane pores we named ceramidosomes. They further suggest that, in addition to their roles in stress-mediated necroptosis, these ceramide-enriched pores also regulate membrane integrity and signaling and might also play a role in D. melanogaster ovary development.
Collapse
Affiliation(s)
- Rose Nganga
- From the Department of Biochemistry and Molecular Biology and.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Natalia Oleinik
- From the Department of Biochemistry and Molecular Biology and.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Jisun Kim
- From the Department of Biochemistry and Molecular Biology and.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Shanmugam Panneer Selvam
- From the Department of Biochemistry and Molecular Biology and.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Ryan De Palma
- From the Department of Biochemistry and Molecular Biology and.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Kristen A Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, Indiana, 46617
| | - Rasesh Y Parikh
- From the Department of Biochemistry and Molecular Biology and
| | - Vamsi Gangaraju
- From the Department of Biochemistry and Molecular Biology and
| | - Yuri Peterson
- the College of Pharmacy/Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, 29425
| | - Mohammed Dany
- From the Department of Biochemistry and Molecular Biology and.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Robert V Stahelin
- the Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907
| | | | | | - Erhard Bieberich
- the Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia 30912, and.,the Department of Physiology, University of Kentucky, Lexington, Kentucky 40506
| | - Besim Ogretmen
- From the Department of Biochemistry and Molecular Biology and .,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425
| |
Collapse
|
43
|
Ameku T, Yoshinari Y, Texada MJ, Kondo S, Amezawa K, Yoshizaki G, Shimada-Niwa Y, Niwa R. Midgut-derived neuropeptide F controls germline stem cell proliferation in a mating-dependent manner. PLoS Biol 2018; 16:e2005004. [PMID: 30248087 PMCID: PMC6152996 DOI: 10.1371/journal.pbio.2005004] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 08/20/2018] [Indexed: 01/21/2023] Open
Abstract
Stem cell maintenance is established by neighboring niche cells that promote stem cell self-renewal. However, it is poorly understood how stem cell activity is regulated by systemic, tissue-extrinsic signals in response to environmental cues and changes in physiological status. Here, we show that neuropeptide F (NPF) signaling plays an important role in the pathway regulating mating-induced germline stem cell (GSC) proliferation in the fruit fly Drosophila melanogaster. NPF expressed in enteroendocrine cells (EECs) of the midgut is released in response to the seminal-fluid protein sex peptide (SP) upon mating. This midgut-derived NPF controls mating-induced GSC proliferation via ovarian NPF receptor (NPFR) activity, which modulates bone morphogenetic protein (BMP) signaling levels in GSCs. Our study provides a molecular mechanism that describes how a gut-derived systemic factor couples stem cell behavior to physiological status, such as mating, through interorgan communication. Communication between different organs is essential to respond quickly to environmental cues or changes in the physiological status of an organism. Recent studies have shown the existence of humoral factors or hormones, which are transported by the circulatory system to different organs and achieve coordination between them. Here, we have analyzed the communication mechanism between organs that regulates proliferation of germline stem cells (GSCs) in the ovary of the fruit fly Drosophila melanogaster. We show that a peptide hormone called neuropeptide F (NPF) is a key player in this process. This peptide is produced in both the brain and the midgut, and, remarkably, we find that only NPF released from the midgut is crucial for controlling post-mating GSC proliferation. Our data suggest that mating stimulates the release of NPF from the endocrine cells of the midgut stimulated by the presence of a seminal peptide. Midgut-derived NPF is then transduced through an NPF-specific G-protein–coupled receptor expressed in the ovary, and this triggers GSC proliferation. Our study identifies an essential interaction between the digestive system and the ovary that regulates the size of stem cell populations in flies depending on mating.
Collapse
Affiliation(s)
- Tomotsune Ameku
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Yuto Yoshinari
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Michael J Texada
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
| | - Shu Kondo
- Genetic Strains Research Center, National Institute of Genetics, Mishima, Japan
| | - Kotaro Amezawa
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Goro Yoshizaki
- Department of Marine Biosciences, Tokyo University of Marine Science and Technology, Tokyo, Japan
| | - Yuko Shimada-Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| | - Ryusuke Niwa
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
44
|
Insulin signaling acts in adult adipocytes via GSK-3β and independently of FOXO to control Drosophila female germline stem cell numbers. Dev Biol 2018; 440:31-39. [PMID: 29729259 DOI: 10.1016/j.ydbio.2018.04.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 04/30/2018] [Accepted: 04/30/2018] [Indexed: 12/14/2022]
Abstract
Tissue-specific stem cells are tied to the nutritional and physiological environment of adult organisms. Adipocytes have key endocrine and nutrient-sensing roles and have emerged as major players in relaying dietary information to regulate other organs. For example, previous studies in Drosophila melanogaster revealed that amino acid sensing as well as diet-dependent metabolic pathways function in adipocytes to influence the maintenance of female germline stem cells (GSCs). How nutrient-sensing pathways acting within adipocytes influence adult stem cell lineages, however, is just beginning to be elucidated. Here, we report that insulin/insulin-like growth factor signaling in adipocytes promotes GSC maintenance, early germline cyst survival, and vitellogenesis. Further, adipocytes use distinct mechanisms downstream of insulin receptor activation to control these aspects of oogenesis, all of which are independent of FOXO. We find that GSC maintenance is modulated by Akt1 through GSK-3β, early germline cyst survival is downstream of adipocyte Akt1 but independent of GSK-3β, and vitellogenesis is regulated through an Akt1-independent pathway in adipocytes. These results indicate that, in addition to employing different types of nutrient sensing, adipocytes can use distinct axes of a single nutrient-sensing pathway to regulate multiple stages of the GSC lineage in the ovary.
Collapse
|
45
|
Kucherenko MM, Shcherbata HR. miRNA targeting and alternative splicing in the stress response - events hosted by membrane-less compartments. J Cell Sci 2018; 131:131/4/jcs202002. [PMID: 29444950 DOI: 10.1242/jcs.202002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stress can be temporary or chronic, and mild or acute. Depending on its extent and severity, cells either alter their metabolism, and adopt a new state, or die. Fluctuations in environmental conditions occur frequently, and such stress disturbs cellular homeostasis, but in general, stresses are reversible and last only a short time. There is increasing evidence that regulation of gene expression in response to temporal stress happens post-transcriptionally in specialized subcellular membrane-less compartments called ribonucleoprotein (RNP) granules. RNP granules assemble through a concentration-dependent liquid-liquid phase separation of RNA-binding proteins that contain low-complexity sequence domains (LCDs). Interestingly, many factors that regulate microRNA (miRNA) biogenesis and alternative splicing are RNA-binding proteins that contain LCDs and localize to stress-induced liquid-like compartments. Consequently, gene silencing through miRNAs and alternative splicing of pre-mRNAs are emerging as crucial post-transcriptional mechanisms that function on a genome-wide scale to regulate the cellular stress response. In this Review, we describe the interplay between these two post-transcriptional processes that occur in liquid-like compartments as an adaptive cellular response to stress.
Collapse
Affiliation(s)
- Mariya M Kucherenko
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany
| | - Halyna R Shcherbata
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Goettingen, Germany
| |
Collapse
|
46
|
Yatsenko AS, Shcherbata HR. Stereotypical architecture of the stem cell niche is spatiotemporally established by miR-125-dependent coordination of Notch and steroid signaling. Development 2018; 145:dev.159178. [PMID: 29361571 PMCID: PMC5818007 DOI: 10.1242/dev.159178] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 01/15/2018] [Indexed: 12/15/2022]
Abstract
Stem cell niches act as signaling platforms that regulate stem cell self-renewal and sustain stem cells throughout life; however, the specific developmental events controlling their assembly are not well understood. Here, we show that during Drosophila ovarian germline stem cell niche formation, the status of Notch signaling in the cell can be reprogrammed. This is controlled via steroid-induced miR-125, which targets a negative regulator of Notch signaling, Tom. Thus, miR-125 acts as a spatiotemporal coordinator between paracrine Notch and endocrine steroid signaling. Moreover, a dual security mechanism for Notch signaling activation exists to ensure the robustness of niche assembly. Particularly, stem cell niche cells can be specified either via lateral inhibition, in which a niche cell precursor acquires Notch signal-sending status randomly, or via peripheral induction, whereby Delta is produced by a specific cell. When one mechanism is perturbed due to mutations, developmental defects or environmental stress, the remaining mechanism ensures that the niche is formed, perhaps abnormally, but still functional. This guarantees that the germline stem cells will have their residence, thereby securing progressive oogenesis and, thus, organism reproduction. Highlighted Article: In Drosophila, the robustness of stem cell niche assembly is safeguarded via a dual mechanism of Notch activation. Cellular Notch status can be reprogrammed by miR-125, which spatiotemporally coordinates paracrine and endocrine signaling.
Collapse
Affiliation(s)
- Andriy S Yatsenko
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| | - Halyna R Shcherbata
- Max Planck Research Group of Gene Expression and Signaling, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany
| |
Collapse
|
47
|
Roy S, Saha TT, Zou Z, Raikhel AS. Regulatory Pathways Controlling Female Insect Reproduction. ANNUAL REVIEW OF ENTOMOLOGY 2018; 63:489-511. [PMID: 29058980 DOI: 10.1146/annurev-ento-020117-043258] [Citation(s) in RCA: 359] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The synthesis of vitellogenin and its uptake by maturing oocytes during egg maturation are essential for successful female reproduction. These events are regulated by the juvenile hormones and ecdysteroids and by the nutritional signaling pathway regulated by neuropeptides. Juvenile hormones act as gonadotropins, regulating vitellogenesis in most insects, but ecdysteroids control this process in Diptera and some Hymenoptera and Lepidoptera. The complex crosstalk between the juvenile hormones, ecdysteroids, and nutritional signaling pathways differs distinctly depending on the reproductive strategies adopted by various insects. Molecular studies within the past decade have revealed much about the relationships among, and the role of, these pathways with respect to regulation of insect reproduction. Here, we review the role of juvenile hormones, ecdysteroids, and nutritional signaling, along with that of microRNAs, in regulating female insect reproduction at the molecular level.
Collapse
Affiliation(s)
- Sourav Roy
- Department of Entomology, Institute for Integrative Genome Biology, and Center for Disease Vector Research, University of California, Riverside, California 92521, USA; , ,
| | - Tusar T Saha
- Department of Entomology, Institute for Integrative Genome Biology, and Center for Disease Vector Research, University of California, Riverside, California 92521, USA; , ,
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Alexander S Raikhel
- Department of Entomology, Institute for Integrative Genome Biology, and Center for Disease Vector Research, University of California, Riverside, California 92521, USA; , ,
| |
Collapse
|
48
|
Banisch TU, Maimon I, Dadosh T, Gilboa L. Escort cells generate a dynamic compartment for germline stem cell differentiation via combined Stat and Erk signalling. Development 2017; 144:1937-1947. [PMID: 28559239 DOI: 10.1242/dev.143727] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 04/21/2017] [Indexed: 01/01/2023]
Abstract
Two different compartments support germline stem cell (GSC) self-renewal and their timely differentiation: the classical niche provides maintenance cues, while a differentiation compartment, formed by somatic escort cells (ECs), is required for proper GSC differentiation. ECs extend long protrusions that invade between tightly packed germ cells, and alternate between encapsulating and releasing them. How ECs achieve this dynamic balance has not been resolved. By combining live imaging and genetic analyses in Drosophila, we have characterised EC shapes and their dynamic changes. We show that germ cell encapsulation by ECs is a communal phenomenon, whereby EC-EC contacts stabilise an extensive meshwork of protrusions. We further show that Signal Transducer and Activator of Transcription (Stat) and Epidermal Growth Factor Receptor (Egfr) signalling sustain EC protrusiveness and flexibility by combinatorially affecting the activity of different RhoGTPases. Our results reveal how a complex signalling network can determine the shape of a cell and its dynamic behaviour. It also explains how the differentiation compartment can establish extensive contacts with germ cells, while allowing a continual posterior movement of differentiating GSC daughters.
Collapse
Affiliation(s)
- Torsten U Banisch
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Iris Maimon
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tali Dadosh
- Electron Microscopy Unit, Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Lilach Gilboa
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
49
|
Wang JB, Lu HL, St. Leger RJ. The genetic basis for variation in resistance to infection in the Drosophila melanogaster genetic reference panel. PLoS Pathog 2017; 13:e1006260. [PMID: 28257468 PMCID: PMC5352145 DOI: 10.1371/journal.ppat.1006260] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/15/2017] [Accepted: 02/24/2017] [Indexed: 01/01/2023] Open
Abstract
Individuals vary extensively in the way they respond to disease but the genetic basis of this variation is not fully understood. We found substantial individual variation in resistance and tolerance to the fungal pathogen Metarhizium anisopliae Ma549 using the Drosophila melanogaster Genetic Reference Panel (DGRP). In addition, we found that host defense to Ma549 was correlated with defense to the bacterium Pseudomonas aeruginosa Pa14, and several previously published DGRP phenotypes including oxidative stress sensitivity, starvation stress resistance, hemolymph glucose levels, and sleep indices. We identified polymorphisms associated with differences between lines in both their mean survival times and microenvironmental plasticity, suggesting that lines differ in their ability to adapt to variable pathogen exposures. The majority of polymorphisms increasing resistance to Ma549 were sex biased, located in non-coding regions, had moderately large effect and were rare, suggesting that there is a general cost to defense. Nevertheless, host defense was not negatively correlated with overall longevity and fecundity. In contrast to Ma549, minor alleles were concentrated in the most Pa14-susceptible as well as the most Pa14-resistant lines. A pathway based analysis revealed a network of Pa14 and Ma549-resistance genes that are functionally connected through processes that encompass phagocytosis and engulfment, cell mobility, intermediary metabolism, protein phosphorylation, axon guidance, response to DNA damage, and drug metabolism. Functional testing with insertional mutagenesis lines indicates that 12/13 candidate genes tested influence susceptibility to Ma549. Many candidate genes have homologs identified in studies of human disease, suggesting that genes affecting variation in susceptibility are conserved across species.
Collapse
Affiliation(s)
- Jonathan B. Wang
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Hsiao-Ling Lu
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
| | - Raymond J. St. Leger
- Department of Entomology, University of Maryland College Park, College Park, Maryland, United States of America
- * E-mail:
| |
Collapse
|
50
|
Ables ET, Drummond-Barbosa D. Steroid Hormones and the Physiological Regulation of Tissue-Resident Stem Cells: Lessons from the Drosophila Ovary. CURRENT STEM CELL REPORTS 2017; 3:9-18. [PMID: 28458991 PMCID: PMC5407287 DOI: 10.1007/s40778-017-0070-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Stem cells respond to local paracrine signals; more recently, however, systemic hormones have also emerged as key regulators of stem cells. This review explores the role of steroid hormones in stem cells, using the Drosophila germline stem cell as a centerpiece for discussion. RECENT FINDINGS Stem cells sense and respond directly and indirectly to steroid hormones, which regulate diverse sets of target genes via interactions with nuclear hormone receptors. Hormone-regulated networks likely integrate the actions of multiple systemic signals to adjust the activity of stem cell lineages in response to changes in physiological status. SUMMARY Hormones are inextricably linked to animal physiology, and can control stem cells and their local niches. Elucidating the molecular mechanisms of hormone signaling in stem cells is essential for our understanding of the fundamental underpinnings of stem cell biology, and for informing new therapeutic interventions against cancers or for regenerative medicine.
Collapse
Affiliation(s)
- Elizabeth T. Ables
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Daniela Drummond-Barbosa
- Department of Biochemistry and Molecular Biology, Division of Reproductive Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|