1
|
Wang Y, Tamori Y. Polyploid Cancer Cell Models in Drosophila. Genes (Basel) 2024; 15:96. [PMID: 38254985 PMCID: PMC10815460 DOI: 10.3390/genes15010096] [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: 11/06/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Cells with an abnormal number of chromosomes have been found in more than 90% of solid tumors, and among these, polyploidy accounts for about 40%. Polyploidized cells most often have duplicate centrosomes as well as genomes, and thus their mitosis tends to promote merotelic spindle attachments and chromosomal instability, which produces a variety of aneuploid daughter cells. Polyploid cells have been found highly resistant to various stress and anticancer therapies, such as radiation and mitogenic inhibitors. In other words, common cancer therapies kill proliferative diploid cells, which make up the majority of cancer tissues, while polyploid cells, which lurk in smaller numbers, may survive. The surviving polyploid cells, prompted by acute environmental changes, begin to mitose with chromosomal instability, leading to an explosion of genetic heterogeneity and a concomitant cell competition and adaptive evolution. The result is a recurrence of the cancer during which the tenacious cells that survived treatment express malignant traits. Although the presence of polyploid cells in cancer tissues has been observed for more than 150 years, the function and exact role of these cells in cancer progression has remained elusive. For this reason, there is currently no effective therapeutic treatment directed against polyploid cells. This is due in part to the lack of suitable experimental models, but recently several models have become available to study polyploid cells in vivo. We propose that the experimental models in Drosophila, for which genetic techniques are highly developed, could be very useful in deciphering mechanisms of polyploidy and its role in cancer progression.
Collapse
Affiliation(s)
| | - Yoichiro Tamori
- Department of Molecular Oncology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| |
Collapse
|
2
|
Cullen G, Gilligan JB, Guhlin JG, Dearden PK. Germline progenitors and oocyte production in the honeybee queen ovary. Genetics 2023; 225:iyad138. [PMID: 37487025 PMCID: PMC10471204 DOI: 10.1093/genetics/iyad138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/26/2023] Open
Abstract
Understanding the reproduction of honeybee queens is crucial to support populations of this economically important insect. Here we examine the structure of the honeybee ovary to determine the nature of the germline progenitors in the ovary. Using a panel of marker genes that mark somatic or germline tissue in other insects we determine which cells in the honeybee ovary are somatic and which germline. We examine patterns of cell division and demonstrate that, unlike Drosophila, there is no evidence of single germline stem cells that provide the germline in honeybees. Germline progenitors are clustered in groups of 8 cells, joined by a polyfusome, and collections of these, in each ovariole, appear to maintain the germline during reproduction. We also show that these 8-cell clusters can divide and that their division occurs such that the numbers of germline progenitors are relatively constant over the reproductive life of queen honeybees. This information helps us to understand the diversity of structures in insect reproduction, and provide information to better support honeybee reproduction.
Collapse
Affiliation(s)
- Georgia Cullen
- Laboratory for Evolution and Development, Biochemistry Department, University of Otago, Dunedin, 9054, Aotearoa-New Zealand
| | - Joshua B Gilligan
- Laboratory for Evolution and Development, Biochemistry Department, University of Otago, Dunedin, 9054, Aotearoa-New Zealand
- Biological Heritage National Science Challenge, Biochemistry Department, University of Otago, Dunedin, 9054, Aotearoa-New Zealand
| | - Joseph G Guhlin
- Laboratory for Evolution and Development, Biochemistry Department, University of Otago, Dunedin, 9054, Aotearoa-New Zealand
- Genomics Aotearoa, Biochemistry Department, University of Otago, Dunedin, 9054, Aotearoa-New Zealand
| | - Peter K Dearden
- Laboratory for Evolution and Development, Biochemistry Department, University of Otago, Dunedin, 9054, Aotearoa-New Zealand
- Biological Heritage National Science Challenge, Biochemistry Department, University of Otago, Dunedin, 9054, Aotearoa-New Zealand
- Genomics Aotearoa, Biochemistry Department, University of Otago, Dunedin, 9054, Aotearoa-New Zealand
| |
Collapse
|
3
|
Tu R, Tang XA, Xu R, Ping Z, Yu Z, Xie T. Gap junction-transported cAMP from the niche controls stem cell progeny differentiation. Proc Natl Acad Sci U S A 2023; 120:e2304168120. [PMID: 37603749 PMCID: PMC10468610 DOI: 10.1073/pnas.2304168120] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 07/22/2023] [Indexed: 08/23/2023] Open
Abstract
The niche has been shown to control stem cell self-renewal in different tissue types and organisms. Recently, a separate niche has been proposed to control stem cell progeny differentiation, called the differentiation niche. However, it remains poorly understood whether and how the differentiation niche directly signals to stem cell progeny to control their differentiation. In the Drosophila ovary, inner germarial sheath (IGS) cells contribute to two separate niche compartments for controlling both germline stem cell (GSC) self-renewal and progeny differentiation. In this study, we show that IGS cells express Inx2 protein, which forms gap junctions (GJs) with germline-specific Zpg protein to control stepwise GSC lineage development, including GSC self-renewal, germline cyst formation, meiotic double-strand DNA break formation, and oocyte specification. Germline-specific Zpg and IGS-specific Inx2 knockdowns cause similar defects in stepwise GSC development. Additionally, secondary messenger cAMP is transported from IGS cells to GSCs and their progeny via GJs to activate PKA signaling for controlling stepwise GSC development. Therefore, this study demonstrates that the niche directly controls GSC progeny differentiation via the GJ-cAMP-PKA signaling axis, which provides important insights into niche control of stem cell differentiation and highlights the importance of GJ-transported cAMP in tissue regeneration. This may represent a general strategy for the niche to control adult stem cell development in various tissue types and organisms since GJs and cAMP are widely distributed.
Collapse
Affiliation(s)
- Renjun Tu
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong Special Administrative Region (SAR), China
| | - Xiaohan Alex Tang
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong Special Administrative Region (SAR), China
| | - Rui Xu
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong Special Administrative Region (SAR), China
| | - Zhaohua Ping
- Stowers Institute for Medical Research, Kansas City, MO64110
| | - Zulin Yu
- Stowers Institute for Medical Research, Kansas City, MO64110
| | - Ting Xie
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong Special Administrative Region (SAR), China
- Stowers Institute for Medical Research, Kansas City, MO64110
| |
Collapse
|
4
|
Dong Z, Pang L, Liu Z, Sheng Y, Li X, Thibault X, Reilein A, Kalderon D, Huang J. Single-cell expression profile of Drosophila ovarian follicle stem cells illuminates spatial differentiation in the germarium. BMC Biol 2023; 21:143. [PMID: 37340484 PMCID: PMC10283321 DOI: 10.1186/s12915-023-01636-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND How stem cell populations are organized and regulated within adult tissues is important for understanding cancer origins and for developing cell replacement strategies. Paradigms such as mammalian gut stem cells and Drosophila ovarian follicle stem cells (FSC) are characterized by population asymmetry, in which stem cell division and differentiation are separately regulated processes. These stem cells behave stochastically regarding their contributions to derivative cells and also exhibit dynamic spatial heterogeneity. Drosophila FSCs provide an excellent model for understanding how a community of active stem cells maintained by population asymmetry is regulated. Here, we use single-cell RNA sequencing to profile the gene expression patterns of FSCs and their immediate derivatives to investigate heterogeneity within the stem cell population and changes associated with differentiation. RESULTS We describe single-cell RNA sequencing studies of a pre-sorted population of cells that include FSCs and the neighboring cell types, escort cells (ECs) and follicle cells (FCs), which they support. Cell-type assignment relies on anterior-posterior (AP) location within the germarium. We clarify the previously determined location of FSCs and use spatially targeted lineage studies as further confirmation. The scRNA profiles among four clusters are consistent with an AP progression from anterior ECs through posterior ECs and then FSCs, to early FCs. The relative proportion of EC and FSC clusters are in good agreement with the prevalence of those cell types in a germarium. Several genes with graded profiles from ECs to FCs are highlighted as candidate effectors of the inverse gradients of the two principal signaling pathways, Wnt and JAK-STAT, that guide FSC differentiation and division. CONCLUSIONS Our data establishes an important resource of scRNA-seq profiles for FSCs and their immediate derivatives that is based on precise spatial location and functionally established stem cell identity, and facilitates future genetic investigation of regulatory interactions guiding FSC behavior.
Collapse
Affiliation(s)
- Zhi Dong
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Lan Pang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Zhiguo Liu
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Yifeng Sheng
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China
| | - Xavier Thibault
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Amy Reilein
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| | - Jianhua Huang
- Institute of Insect Sciences, Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
5
|
Galasso A, Xu DC, Hill C, Iakovleva D, Stefana MI, Baena‐Lopez LA. Non-apoptotic caspase activation ensures the homeostasis of ovarian somatic stem cells. EMBO Rep 2023; 24:e51716. [PMID: 37039000 PMCID: PMC10240206 DOI: 10.15252/embr.202051716] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/22/2023] [Accepted: 03/14/2023] [Indexed: 04/12/2023] Open
Abstract
Current evidence has associated caspase activation with the regulation of basic cellular functions without causing apoptosis. Malfunction of non-apoptotic caspase activities may contribute to specific neurological disorders, metabolic diseases, autoimmune conditions and cancers. However, our understanding of non-apoptotic caspase functions remains limited. Here, we show that non-apoptotic caspase activation prevents the intracellular accumulation of the Patched receptor in autophagosomes and the subsequent Patched-dependent induction of autophagy in Drosophila follicular stem cells. These events ultimately sustain Hedgehog signalling and the physiological properties of ovarian somatic stem cells and their progeny under moderate thermal stress. Importantly, our key findings are partially conserved in ovarian somatic cells of human origin. These observations attribute to caspases a pro-survival role under certain cellular conditions.
Collapse
Affiliation(s)
- Alessia Galasso
- Faculty of Medicine CentreImperial College London, South Kensington CampusLondonUK
| | - Derek Cui Xu
- Sir William Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Claire Hill
- School of Medicine, Dentistry and Biomedical SciencesQueen's University Belfast MedicineBelfastUK
| | - Daria Iakovleva
- Center for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | | | | |
Collapse
|
6
|
Gao J, Gao Y, Xiao G. The expression of Catsup in escort cells affects Drosophila ovarian stem cell niche establishment and germline stem cells self-renewal via Notch signaling. Biochem Biophys Res Commun 2023; 641:1-9. [PMID: 36516479 DOI: 10.1016/j.bbrc.2022.11.101] [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: 11/20/2022] [Accepted: 11/30/2022] [Indexed: 12/07/2022]
Abstract
Stem cell niche provides extrinsic signals to maintain stem cell renewal or initiate cell differentiation. Drosophila niche is composed of somatic terminal filament cells, cap cells and escort cells. However, the underlying mechanism for the development of stem cell niche remains largely unclear. Here we found that the expression of a zinc transporter Catsup is essential for ovary morphogenesis. Catsup knockdown in escort cells results in defects of niche establishment and germline stem cells self-renewal. These defects could be modified by altered expression of genes involved in zinc metabolism or intervention of dietary zinc levels. Further studies indicated that Catsup RNAi affected adult ovary morphogenesis by suppressing Notch signaling. Lastly, we demonstrated that the defects of Catsup RNAi could be restored by overexpression of heat shock cognate protein 70 (Hsc70). These findings expand our understanding of the mechanisms controlling adult oogenesis and niche establishment in Drosophila.
Collapse
Affiliation(s)
- Jiajia Gao
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, 230009, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Yan Gao
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, 230009, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Guiran Xiao
- China Light Industry Key Laboratory of Meat Microbial Control and Utilization, Hefei University of Technology, Hefei, 230009, China; Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China.
| |
Collapse
|
7
|
Wang QQ, You DD, Liu JL. Cytoophidia Maintain the Integrity of Drosophila Follicle Epithelium. Int J Mol Sci 2022; 23:ijms232315282. [PMID: 36499609 PMCID: PMC9740582 DOI: 10.3390/ijms232315282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
CTP synthase (CTPS) forms a filamentous structure termed the cytoophidium in all three domains of life. The female reproductive system of Drosophila is an excellent model for studying the physiological function of cytoophidia. Here, we use CTPSH355A, a point mutation that destroys the cytoophidium-forming ability of CTPS, to explore the in vivo function of cytoophidia. In CTPSH355A egg chambers, we observe the ingression and increased heterogeneity of follicle cells. In addition, we find that the cytoophidium-forming ability of CTPS, rather than the protein level, is the cause of the defects observed in CTPSH355A mutants. To sum up, our data indicate that cytoophidia play an important role in maintaining the integrity of follicle epithelium.
Collapse
Affiliation(s)
- Qiao-Qi Wang
- School of Life Science and Technology, Shanghai Technology University, Shanghai 201210, China
| | - Dong-Dong You
- School of Life Science and Technology, Shanghai Technology University, Shanghai 201210, China
| | - Ji-Long Liu
- School of Life Science and Technology, Shanghai Technology University, Shanghai 201210, China
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Correspondence: or
| |
Collapse
|
8
|
Bostock MP, Prasad AR, Donoghue A, Fernandes VM. Photoreceptors generate neuronal diversity in their target field through a Hedgehog morphogen gradient in Drosophila. eLife 2022; 11:78093. [PMID: 36004721 PMCID: PMC9507128 DOI: 10.7554/elife.78093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Defining the origin of neuronal diversity is a major challenge in developmental neurobiology. The Drosophila visual system is an excellent paradigm to study how cellular diversity is generated. Photoreceptors from the eye disc grow their axons into the optic lobe and secrete Hedgehog (Hh) to induce the lamina, such that for every unit eye there is a corresponding lamina unit made up of post-mitotic precursors stacked into columns. Each differentiated column contains five lamina neuron types (L1-L5), making it the simplest neuropil in the optic lobe, yet how this diversity is generated was unknown. Here, we found that Hh pathway activity is graded along the distal-proximal axis of lamina columns and further determined that this gradient in pathway activity arises from a gradient of Hh ligand. We manipulated Hh pathway activity cell-autonomously in lamina precursors and non-cell autonomously by inactivating the Hh ligand, and by knocking it down in photoreceptors. These manipulations showed that different thresholds of activity specify unique cell identities, with more proximal cell types specified in response to progressively lower Hh levels. Thus, our data establish that Hh acts as a morphogen to pattern the lamina. Although, this is the first such report during Drosophila nervous system development, our work uncovers a remarkable similarity with the vertebrate neural tube, which is patterned by Sonic Hedgehog. Altogether, we show that differentiating neurons can regulate the neuronal diversity of their distant target fields through morphogen gradients.
Collapse
Affiliation(s)
- Matthew P Bostock
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Anadika R Prasad
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Alicia Donoghue
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Vilaiwan M Fernandes
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
9
|
Diegmiller R, Nunley H, Shvartsman SY, Imran Alsous J. Quantitative models for building and growing fated small cell networks. Interface Focus 2022; 12:20210082. [PMID: 35865502 PMCID: PMC9184967 DOI: 10.1098/rsfs.2021.0082] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/31/2022] [Indexed: 02/07/2023] Open
Abstract
Small cell clusters exhibit numerous phenomena typically associated with complex systems, such as division of labour and programmed cell death. A conserved class of such clusters occurs during oogenesis in the form of germline cysts that give rise to oocytes. Germline cysts form through cell divisions with incomplete cytokinesis, leaving cells intimately connected through intercellular bridges that facilitate cyst generation, cell fate determination and collective growth dynamics. Using the well-characterized Drosophila melanogaster female germline cyst as a foundation, we present mathematical models rooted in the dynamics of cell cycle proteins and their interactions to explain the generation of germline cell lineage trees (CLTs) and highlight the diversity of observed CLT sizes and topologies across species. We analyse competing models of symmetry breaking in CLTs to rationalize the observed dynamics and robustness of oocyte fate specification, and highlight remaining gaps in knowledge. We also explore how CLT topology affects cell cycle dynamics and synchronization and highlight mechanisms of intercellular coupling that underlie the observed collective growth patterns during oogenesis. Throughout, we point to similarities across organisms that warrant further investigation and comment on the extent to which experimental and theoretical findings made in model systems extend to other species.
Collapse
Affiliation(s)
- Rocky Diegmiller
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Hayden Nunley
- Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Stanislav Y. Shvartsman
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA,Department of Molecular Biology, Princeton University, Princeton, NJ, USA,Flatiron Institute, Simons Foundation, New York, NY, USA
| | | |
Collapse
|
10
|
Chen TA, Lin KY, Yang SM, Tseng CY, Wang YT, Lin CH, Luo L, Cai Y, Hsu HJ. Canonical Wnt Signaling Promotes Formation of Somatic Permeability Barrier for Proper Germ Cell Differentiation. Front Cell Dev Biol 2022; 10:877047. [PMID: 35517512 PMCID: PMC9062081 DOI: 10.3389/fcell.2022.877047] [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: 02/16/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022] Open
Abstract
Morphogen-mediated signaling is critical for proper organ development and stem cell function, and well-characterized mechanisms spatiotemporally limit the expression of ligands, receptors, and ligand-binding cell-surface glypicans. Here, we show that in the developing Drosophila ovary, canonical Wnt signaling promotes the formation of somatic escort cells (ECs) and their protrusions, which establish a physical permeability barrier to define morphogen territories for proper germ cell differentiation. The protrusions shield germ cells from Dpp and Wingless morphogens produced by the germline stem cell (GSC) niche and normally only received by GSCs. Genetic disruption of EC protrusions allows GSC progeny to also receive Dpp and Wingless, which subsequently disrupt germ cell differentiation. Our results reveal a role for canonical Wnt signaling in specifying the ovarian somatic cells necessary for germ cell differentiation. Additionally, we demonstrate the morphogen-limiting function of this physical permeability barrier, which may be a common mechanism in other organs across species.
Collapse
Affiliation(s)
- Ting-An Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Kun-Yang Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Shun-Min Yang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- Faculty of Science, University of South Bohemia, České Budějovice, Czechia
| | - Chen-Yuan Tseng
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yu-Ting Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chi-Hung Lin
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Lichao Luo
- Temasek Life Science Laboratory, National University of Singapore, Singapore, Singapore
| | - Yu Cai
- Temasek Life Science Laboratory, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Hwei-Jan Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
- *Correspondence: Hwei-Jan Hsu,
| |
Collapse
|
11
|
Dunipace L, Newcomb S, Stathopoulos A. brinker levels regulated by a promoter proximal element support germ cell homeostasis. Development 2022; 149:274023. [PMID: 35037688 PMCID: PMC8918798 DOI: 10.1242/dev.199890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022]
Abstract
ABSTRACT
A limited BMP signaling range in the stem cell niche of the ovary protects against germ cell tumors and promotes germ cell homeostasis. The canonical repressor of BMP signaling in both the Drosophila embryo and wing disc is the transcription factor Brinker (Brk), yet the expression and potential role of Brk in the germarium has not previously been described. Here, we find that brk expression requires a promoter-proximal element (PPE) to support long-distance enhancer action as well as to drive expression in the germarium. Furthermore, PPE subdomains have different activities; in particular, the proximal portion acts as a damper to regulate brk levels precisely. Using PPE mutants as well as tissue-specific RNA interference and overexpression, we show that altering brk expression within either the soma or the germline affects germ cell homeostasis. Remarkably, we find that Decapentaplegic (Dpp), the main BMP ligand and canonical antagonist of Brk, is upregulated by Brk in the escort cells of the germarium, demonstrating that Brk can positively regulate this pathway.
Collapse
Affiliation(s)
- Leslie Dunipace
- Division of Biology, California Institute of Technology, 1200 East California Boulevard, MC114-96, Pasadena, CA 91125, USA
| | - Susan Newcomb
- Division of Biology, California Institute of Technology, 1200 East California Boulevard, MC114-96, Pasadena, CA 91125, USA
| | - Angelike Stathopoulos
- Division of Biology, California Institute of Technology, 1200 East California Boulevard, MC114-96, Pasadena, CA 91125, USA
| |
Collapse
|
12
|
Abstract
A simple, universal and fundamental definition of adult stem cell communities is proposed. Key principles of cell lineage methods for defining adult stem cell numbers, locations and behaviors are critically evaluated, emphasizing the imperatives of capturing the full spectrum of individual stem cell behaviors, examining a variety of experimental time periods and avoiding unwarranted assumptions. The focus is first on defining fundamentals and then addresses stem cell heterogeneity, potential hierarchies and how individual cells serve the function of a stem cell community.
Collapse
|
13
|
Tatapudy S, Peralta J, Nystul T. Distinct roles of Bendless in regulating FSC niche competition and daughter cell differentiation. Development 2021; 148:dev199630. [PMID: 35020878 PMCID: PMC8645206 DOI: 10.1242/dev.199630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/13/2021] [Indexed: 04/05/2024]
Abstract
A major goal in the study of adult stem cells is to understand how cell fates are specified at the proper time and place to facilitate tissue homeostasis. Here, we found that an E2 ubiquitin ligase, Bendless (Ben), has multiple roles in the Drosophila ovarian epithelial follicle stem cell (FSC) lineage. First, Ben is part of the JNK signaling pathway, and we found that it, as well as other JNK pathway genes, are essential for differentiation of FSC daughter cells. Our data suggest that JNK signaling promotes differentiation by suppressing the activation of the EGFR effector, ERK. Also, we found that loss of ben, but not the JNK kinase hemipterous, resulted in an upregulation of hedgehog signaling, increased proliferation and increased niche competition. Lastly, we demonstrate that the hypercompetition phenotype caused by loss of ben is suppressed by decreasing the rate of proliferation or knockdown of the hedgehog pathway effector, Smoothened (Smo). Taken together, our findings reveal a new layer of regulation in which a single gene influences cell signaling at multiple stages of differentiation in the early FSC lineage.
Collapse
Affiliation(s)
| | | | - Todd Nystul
- Department of Anatomy and Department of OB/Gyn-RS, University of California, San Francisco, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| |
Collapse
|
14
|
Reilein A, Kogan HV, Misner R, Park KS, Kalderon D. Adult stem cells and niche cells segregate gradually from common precursors that build the adult Drosophila ovary during pupal development. eLife 2021; 10:69749. [PMID: 34590579 PMCID: PMC8536258 DOI: 10.7554/elife.69749] [Citation(s) in RCA: 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: 04/24/2021] [Accepted: 09/29/2021] [Indexed: 12/31/2022] Open
Abstract
Production of proliferative follicle cells (FCs) and quiescent escort cells (ECs) by follicle stem cells (FSCs) in adult Drosophila ovaries is regulated by niche signals from anterior (cap cells, ECs) and posterior (polar FCs) sources. Here we show that ECs, FSCs, and FCs develop from common pupal precursors, with different fates acquired by progressive separation of cells along the AP axis and a graded decline in anterior cell proliferation. ECs, FSCs, and most FCs derive from intermingled cell (IC) precursors interspersed with germline cells. Precursors also accumulate posterior to ICs before engulfing a naked germline cyst projected out of the germarium to form the first egg chamber and posterior polar FC signaling center. Thus, stem and niche cells develop in appropriate numbers and spatial organization through regulated proliferative expansion together with progressive establishment of spatial signaling cues that guide adult cell behavior, rather than through rigid early specification events.
Collapse
Affiliation(s)
- Amy Reilein
- Department of Biological Sciences, Columbia University, New York, United States
| | - Helen V Kogan
- Department of Biological Sciences, Columbia University, New York, United States
| | - Rachel Misner
- Department of Biological Sciences, Columbia University, New York, United States
| | - Karen Sophia Park
- Department of Biological Sciences, Columbia University, New York, United States
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, United States
| |
Collapse
|
15
|
Slaidina M, Gupta S, Banisch TU, Lehmann R. A single-cell atlas reveals unanticipated cell type complexity in Drosophila ovaries. Genome Res 2021; 31:1938-1951. [PMID: 34389661 DOI: 10.1101/gr.274340.120] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 08/09/2021] [Indexed: 11/24/2022]
Abstract
Organ function relies on the spatial organization and functional coordination of numerous cell types. The Drosophila ovary is a widely used model system to study the cellular activities underlying organ function, including stem cell regulation, cell signaling and epithelial morphogenesis. However, the relative paucity of cell type-specific reagents hinders investigation of molecular functions at the appropriate cellular resolution. Here, we used single-cell RNA sequencing to characterize all cell types of the stem cell compartment and early follicles of the Drosophila ovary. We computed transcriptional signatures and identified specific markers for nine states of germ cell differentiation, and 23 somatic cell types and subtypes. We uncovered an unanticipated diversity of escort cells, the somatic cells that directly interact with differentiating germline cysts. Three escort cell subtypes reside in discrete anatomical positions, and express distinct sets of secreted and transmembrane proteins, suggesting that diverse micro-environments support the progressive differentiation of germ cells. Finally, we identified 17 follicle cell subtypes, and characterized their transcriptional profiles. Altogether, we provide a comprehensive resource of gene expression, cell type-specific markers, spatial coordinates and functional predictions for 34 ovarian cell types and subtypes.
Collapse
Affiliation(s)
| | - Selena Gupta
- Skirball Institute, NYU Grossman School of Medicine
| | | | | |
Collapse
|
16
|
Beachum AN, Whitehead KM, McDonald SI, Phipps DN, Berghout HE, Ables ET. Orphan nuclear receptor ftz-f1 (NR5A3) promotes egg chamber survival in the Drosophila ovary. G3-GENES GENOMES GENETICS 2021; 11:6114459. [PMID: 33693603 PMCID: PMC8022936 DOI: 10.1093/g3journal/jkab003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/30/2020] [Indexed: 11/12/2022]
Abstract
Gamete production in mammals and insects is controlled by cell signaling pathways that facilitate communication between germ cells and somatic cells. Nuclear receptor signaling is a key mediator of many aspects of reproduction, including gametogenesis. For example, the NR5A subfamily of nuclear receptors is essential for gonad development and sex steroid production in mammals. Despite the original identification of the NR5A subfamily in the model insect Drosophila melanogaster, it has been unclear whether Drosophila NR5A receptors directly control oocyte production. Ftz-f1 is expressed throughout the ovary, including in germline stem cells, germline cysts, and several populations of somatic cells. We show that ftz-f1 is required in follicle cells prior to stage 10 to promote egg chamber survival at the mid-oogenesis checkpoint. Our data suggest that egg chamber death in the absence of ftz-f1 is due, at least in part, to failure of follicle cells to exit the mitotic cell cycle or failure to accumulate oocyte-specific factors in the germline. Taken together, these results show that, as in mammals, the NR5A subfamily promotes maximal reproductive output in Drosophila. Our data underscore the importance of nuclear receptors in the control of reproduction and highlight the utility of Drosophila oogenesis as a key model for unraveling the complexity of nuclear receptor signaling in gametogenesis.
Collapse
Affiliation(s)
- Allison N Beachum
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | | | | | - Daniel N Phipps
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Hanna E Berghout
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
| | - Elizabeth T Ables
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
- Corresponding author: Department of Biology, East Carolina University, 1001 E. 10th St., Mailstop 551, 553 Science & Technology Building, Greenville, NC 27858, USA.
| |
Collapse
|
17
|
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
|
18
|
Wang X, LaFever KS, Waghmare I, Page-McCaw A. Extracellular spreading of Wingless is required for Drosophila oogenesis. PLoS Genet 2021; 17:e1009469. [PMID: 33798197 PMCID: PMC8046344 DOI: 10.1371/journal.pgen.1009469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/14/2021] [Accepted: 03/05/2021] [Indexed: 11/18/2022] Open
Abstract
Recent studies have investigated whether the Wnt family of extracellular ligands can signal at long range, spreading from their source and acting as morphogens, or whether they signal only in a juxtacrine manner to neighboring cells. The original evidence for long-range Wnt signaling arose from studies of Wg, a Drosophila Wnt protein, which patterns the wing disc over several cell diameters from a central source of Wg ligand. However, the requirement of long-range Wg for patterning was called into question when it was reported that replacing the secreted protein Wg with a membrane-tethered version, NRT-Wg, results in flies with normally patterned wings. We and others previously reported that Wg spreads in the ovary about 50 μm or 5 cell diameters, from the cap cells to the follicle stem cells (FSCs) and that Wg stimulates FSC proliferation. We used the NRT-wg flies to analyze the consequence of tethering Wg to the cap cells. NRT-wg homozygous flies are sickly, but we found that hemizygous NRT-wg/null flies, carrying only one copy of tethered Wingless, were significantly healthier. Despite their overall improved health, these hemizygous flies displayed dramatic reductions in fertility and in FSC proliferation. Further, FSC proliferation was nearly undetectable when the wg locus was converted to NRT-wg only in adults, and the resulting germarium phenotype was consistent with a previously reported wg loss-of-function phenotype. We conclude that Wg protein spreads from its source cells in the germarium to promote FSC proliferation.
Collapse
Affiliation(s)
- Xiaoxi Wang
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Kimberly S. LaFever
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Indrayani Waghmare
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology, Program in Developmental Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| |
Collapse
|
19
|
Sadanandappa MK, Sathyanarayana SH, Kondo S, Bosco G. Neuropeptide F signaling regulates parasitoid-specific germline development and egg-laying in Drosophila. PLoS Genet 2021; 17:e1009456. [PMID: 33770070 PMCID: PMC8026082 DOI: 10.1371/journal.pgen.1009456] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 04/07/2021] [Accepted: 03/01/2021] [Indexed: 01/08/2023] Open
Abstract
Drosophila larvae and pupae are at high risk of parasitoid infection in nature. To circumvent parasitic stress, fruit flies have developed various survival strategies, including cellular and behavioral defenses. We show that adult Drosophila females exposed to the parasitic wasps, Leptopilina boulardi, decrease their total egg-lay by deploying at least two strategies: Retention of fully developed follicles reduces the number of eggs laid, while induction of caspase-mediated apoptosis eliminates the vitellogenic follicles. These reproductive defense strategies require both visual and olfactory cues, but not the MB247-positive mushroom body neuronal function, suggesting a novel mode of sensory integration mediates reduced egg-laying in the presence of a parasitoid. We further show that neuropeptide F (NPF) signaling is necessary for both retaining matured follicles and activating apoptosis in vitellogenic follicles. Whereas previous studies have found that gut-derived NPF controls germ stem cell proliferation, we show that sensory-induced changes in germ cell development specifically require brain-derived NPF signaling, which recruits a subset of NPFR-expressing cell-types that control follicle development and retention. Importantly, we found that reduced egg-lay behavior is specific to parasitic wasps that infect the developing Drosophila larvae, but not the pupae. Our findings demonstrate that female fruit flies use multimodal sensory integration and neuroendocrine signaling via NPF to engage in parasite-specific cellular and behavioral survival strategies.
Collapse
Affiliation(s)
- Madhumala K. Sadanandappa
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Shivaprasad H. Sathyanarayana
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - Shu Kondo
- Invertebrate Genetics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Giovanni Bosco
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
Zhu G, Gonzalez LE, Lin H. Piwi in the stem cell niche regulates nurse cell number and oocyte specification. MICROPUBLICATION BIOLOGY 2020; 2020:10.17912/micropub.biology.000327. [PMID: 33274324 PMCID: PMC7704254 DOI: 10.17912/micropub.biology.000327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/23/2020] [Accepted: 11/09/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Gina Zhu
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06519, USA
- Yale College, New Haven, CT 06511, USA
| | - Lauren E Gonzalez
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06519, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT 06519, USA
| | - Haifan Lin
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06519, USA
- Department of Genetics, Yale School of Medicine, New Haven, CT 06519, USA
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
23
|
A single-cell atlas and lineage analysis of the adult Drosophila ovary. Nat Commun 2020; 11:5628. [PMID: 33159074 PMCID: PMC7648648 DOI: 10.1038/s41467-020-19361-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/08/2020] [Indexed: 01/05/2023] Open
Abstract
The Drosophila ovary is a widely used model for germ cell and somatic tissue biology. Here we use single-cell RNA-sequencing (scRNA-seq) to build a comprehensive cell atlas of the adult Drosophila ovary that contains transcriptional profiles for every major cell type in the ovary, including the germline stem cells and their niche cells, follicle stem cells, and previously undescribed subpopulations of escort cells. In addition, we identify Gal4 lines with specific expression patterns and perform lineage tracing of subpopulations of escort cells and follicle cells. We discover that a distinct subpopulation of escort cells is able to convert to follicle stem cells in response to starvation or upon genetic manipulation, including knockdown of escargot, or overactivation of mTor or Toll signalling.
Collapse
|
24
|
Abstract
Precise genetic manipulation of specific cell types or tissues to pinpoint gene function requirement is a critical step in studies aimed at unraveling the intricacies of organismal physiology. Drosophila researchers heavily rely on the UAS/Gal4/Gal80 system for tissue-specific manipulations; however, it is often unclear whether the reported Gal4 expression patterns are indeed specific to the tissue of interest such that experimental results are not confounded by secondary sites of Gal4 expression. Here, we surveyed the expression patterns of commonly used Gal4 drivers in adult Drosophila female tissues under optimal conditions and found that multiple drivers have unreported secondary sites of expression beyond their published cell type/tissue expression pattern. These results underscore the importance of thoroughly characterizing Gal4 tools as part of a rigorous experimental design that avoids potential misinterpretation of results as we strive for understanding how the function of a specific gene/pathway in one tissue contributes to whole-body physiology.
Collapse
|
25
|
Melamed D, Kalderon D. Opposing JAK-STAT and Wnt signaling gradients define a stem cell domain by regulating differentiation at two borders. eLife 2020; 9:61204. [PMID: 33135631 PMCID: PMC7695452 DOI: 10.7554/elife.61204] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Many adult stem cell communities are maintained by population asymmetry, where stochastic behaviors of multiple individual cells collectively result in a balance between stem cell division and differentiation. We investigated how this is achieved for Drosophila Follicle Stem Cells (FSCs) by spatially-restricted niche signals. FSCs produce transit-amplifying Follicle Cells (FCs) from their posterior face and quiescent Escort Cells (ECs) to their anterior. We show that JAK-STAT pathway activity, which declines from posterior to anterior, dictates the pattern of divisions over the FSC domain, promotes more posterior FSC locations and conversion to FCs, while opposing EC production. Wnt pathway activity declines from the anterior, promotes anterior FSC locations and EC production, and opposes FC production. The pathways combine to define a stem cell domain through concerted effects on FSC differentiation to ECs and FCs at either end of opposing signaling gradients, and impose a pattern of proliferation that matches derivative production.
Collapse
Affiliation(s)
- David Melamed
- Department of Biological Sciences, Columbia University, New York, United States
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, United States
| |
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
|
Waghmare I, Wang X, Page-McCaw A. Dally-like protein sequesters multiple Wnt ligands in the Drosophila germarium. Dev Biol 2020; 464:88-102. [PMID: 32473955 DOI: 10.1016/j.ydbio.2020.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/02/2020] [Accepted: 05/11/2020] [Indexed: 12/20/2022]
Abstract
Cells in multicellular organisms rely on secreted ligands for development and morphogenesis. Several mechanisms modulate the availability and distribution of secreted ligands, determining their ability to signal locally and at long range from their source. One of these mechanisms is Dally-like protein (Dlp), a cell-surface glypican that exhibits biphasic functions in Drosophila wing discs, promoting Wg signaling at long-range from Wg source cells and inhibiting Wg signaling near source cells. In the germarium at the tip of the ovary, Dlp promotes long-range distribution of Wg from cap cells to follicle stem cells. However, the germarium also expresses other Wnts - Wnt2, Wnt4, and Wnt6 - that function locally in escort cells to promote oogenesis. Whether and how local functions of these Wnts are regulated remains unknown. Here we show that the dlp overexpression phenotype is multifaceted and phenocopies multiple Wnt loss-of-function phenotypes. Each aspect of dlp overexpression phenotype is suppressed by co-expression of individual Wnts, and the suppression pattern exhibited by each Wnt suggests that Wnts have functional specificity in the germarium. Further, dlp knockdown phenocopies Wnt gain-of-function phenotypes. Together these data show that Dlp inhibits the functions of each Wnt. All four Wnts co-immunoprecipitate with Dlp in S2R+ cells, suggesting that in the germarium, Dlp sequesters Wnts to inhibit local paracrine Wnt signaling. Our results indicate that Dlp modulates the availability of multiple extracellular Wnts for local paracrine Wnt signaling in the germarium.
Collapse
Affiliation(s)
- Indrayani Waghmare
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| | - Xiaoxi Wang
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| |
Collapse
|
28
|
Tu R, Duan B, Song X, Xie T. Dlp-mediated Hh and Wnt signaling interdependence is critical in the niche for germline stem cell progeny differentiation. SCIENCE ADVANCES 2020; 6:eaaz0480. [PMID: 32426496 PMCID: PMC7220319 DOI: 10.1126/sciadv.aaz0480] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 02/28/2020] [Indexed: 05/04/2023]
Abstract
Although multiple signaling pathways work synergistically in various niches to control stem cell self-renewal and differentiation, it remains poorly understood how they cooperate with one another molecularly. In the Drosophila ovary, Hh and Wnt pathways function in the niche to promote germline stem cell (GSC) progeny differentiation. Here, we show that glypican Dlp-mediated Hh and Wnt signaling interdependence operates in the niche to promote GSC progeny differentiation by preventing BMP signaling. Hh/Wnt-mediated dlp repression is essential for their signaling interdependence in niche cells and for GSC progeny differentiation by preventing BMP signaling. Mechanistically, Hh and Wnt downstream transcription factors directly bind to the same dlp regulatory region and recruit corepressors composed of transcription factor Croc and Egg/H3K9 trimethylase to repress Dlp expression. Therefore, our study reveals a novel mechanism for Hh/Wnt signaling-mediated direct dlp repression and a novel regulatory mechanism for Dlp-mediated Hh/Wnt signaling interdependence in the GSC differentiation niche.
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
| | - 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
- Corresponding author.
| |
Collapse
|
29
|
Ishibashi JR, Taslim TH, Ruohola-Baker H. Germline stem cell aging in the Drosophila ovary. CURRENT OPINION IN INSECT SCIENCE 2020; 37:57-62. [PMID: 32120010 DOI: 10.1016/j.cois.2019.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/12/2019] [Accepted: 11/16/2019] [Indexed: 06/10/2023]
Abstract
The age-related decline of adult stem cells leads to loss of tissue homeostasis and contributes to organismal aging. Though the phenotypic hallmarks of aging are well-characterized at the organ or tissue level, the molecular processes that govern stem cell aging remain unclear. This review seeks to highlight recent research in stem cell aging in the Drosophila ovary and connect the discoveries in the fly to ongoing questions in stem cell aging.
Collapse
Affiliation(s)
- Julien Roy Ishibashi
- Department of Biochemistry, Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican St, Seattle, WA 98109, United States
| | - Tommy Henry Taslim
- Department of Biochemistry, Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican St, Seattle, WA 98109, United States
| | - Hannele Ruohola-Baker
- Department of Biochemistry, Institute for Stem Cell and Regenerative Medicine, University of Washington, 850 Republican St, Seattle, WA 98109, United States.
| |
Collapse
|
30
|
Rust K, Nystul T. Signal transduction in the early Drosophila follicle stem cell lineage. CURRENT OPINION IN INSECT SCIENCE 2020; 37:39-48. [PMID: 32087562 PMCID: PMC7155752 DOI: 10.1016/j.cois.2019.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/13/2019] [Accepted: 11/16/2019] [Indexed: 05/08/2023]
Abstract
The follicle stem cell (FSC) lineage in the Drosophila ovary is a highly informative model of in vivo epithelial stem cell biology. Studies over the past 30 years have identified roles for every major signaling pathway in the early FSC lineage. These pathways regulate a wide variety of cell behaviors, including self-renewal, proliferation, survival and differentiation. Studies of cell signaling in the follicle epithelium have provided new insights into how these cell behaviors are coordinated within an epithelial stem cell lineage and how signaling pathways interact with each other in the native, in vivo context of a living tissue. Here, we review these studies, with a particular focus on how these pathways specify differences between the FSCs and their daughter cells. We also describe common themes that have emerged from these studies, and highlight new research directions that have been made possible by the detailed understanding of the follicle epithelium.
Collapse
|
31
|
Lobo-Pecellín M, Marín-Menguiano M, González-Reyes A. mastermind regulates niche ageing independently of the Notch pathway in the Drosophila ovary. Open Biol 2019; 9:190127. [PMID: 31744422 PMCID: PMC6893403 DOI: 10.1098/rsob.190127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/21/2019] [Indexed: 01/09/2023] Open
Abstract
Proper stem cell activity in tissues ensures the correct balance between proliferation and differentiation, thus allowing tissue homeostasis and repair. The Drosophila ovary develops well-defined niches that contain on average 2-4 germline stem cells (GSCs), whose maintenance depends on systemic signals and local factors. A known player in the decline of tissue homeostasis is ageing, which correlates with the waning of resident stem cell populations. In Drosophila, ovaries from old females contain fewer GSCs than those from young flies. We isolated niche cells of aged ovaries, performed a transcriptomic analysis and identified mastermind (mam) as a factor for Drosophila ovarian niche functionality during ageing. We show that mam is upregulated in aged niche cells and that we can induce premature GSC loss by overexpressing mam in otherwise young niche cells. High mam levels in niche cells induce reduced Hedgehog amounts, a decrease in cadherin levels and a likely increase in reactive oxygen species, three scenarios known to provoke GSC loss. Mam is a canonical co-activator of the Notch pathway in many Drosophila tissues. However, we present evidence to support a Notch-independent role for mam in the ovarian germline niche.
Collapse
Affiliation(s)
| | | | - Acaimo González-Reyes
- Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Carretera de Utrera km 1, 41013 Sevilla, Spain
| |
Collapse
|
32
|
Kim-Yip RP, Nystul TG. Wingless promotes EGFR signaling in follicle stem cells to maintain self-renewal. Development 2018; 145:dev.168716. [PMID: 30389852 DOI: 10.1242/dev.168716] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022]
Abstract
Adult stem cell niche boundaries must be precisely maintained to facilitate the segregation of stem cell and daughter cell fates. However, the mechanisms that govern this process in epithelial tissues are not fully understood. In this study, we investigated the relationship between two signals, Wnt and EGFR, that are necessary for self-renewal of the epithelial follicle stem cells (FSCs) in the Drosophila ovary, but must be downregulated in cells that have exited the niche to allow for differentiation. We found that Wingless produced by inner germarial sheath (IGS) cells acts over a short distance to activate Wnt signaling in FSCs, and that movement across the FSC niche boundary is limited. In addition, we show that Wnt signaling functions genetically upstream of EGFR signaling by activating the expression of the EGFR ligand, Spitz, and that constitutive activation of EGFR partially rescues the self-renewal defect caused by loss of Wnt signaling. Collectively, our findings support a model in which the Wnt and EGFR pathways operate in a signaling hierarchy to promote FSC self-renewal.
Collapse
Affiliation(s)
- Rebecca P Kim-Yip
- Center for Reproductive Sciences, Departments of Anatomy and OB/GYN-RS, University of California, San Francisco, CA 94143-0452, USA
| | - Todd G Nystul
- Center for Reproductive Sciences, Departments of Anatomy and OB/GYN-RS, University of California, San Francisco, CA 94143-0452, USA
| |
Collapse
|
33
|
Drosophila Glypicans Regulate Follicle Stem Cell Maintenance and Niche Competition. Genetics 2018; 209:537-549. [PMID: 29632032 DOI: 10.1534/genetics.118.300839] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/04/2018] [Indexed: 01/06/2023] Open
Abstract
Adult stem cells reside in specialized microenvironments called niches, which provide signals for stem cells to maintain their undifferentiated and self-renewing state. To maintain stem cell quality, several types of stem cells are known to be regularly replaced by progenitor cells through niche competition. However, the cellular and molecular bases for stem cell competition for niche occupancy are largely unknown. Here, we show that two Drosophila members of the glypican family of heparan sulfate proteoglycans (HSPGs), Dally and Dally-like (Dlp), differentially regulate follicle stem cell (FSC) maintenance and competitiveness for niche occupancy. Lineage analyses of glypican mutant FSC clones showed that dally is essential for normal FSC maintenance. In contrast, dlp is a hypercompetitive mutation: dlp mutant FSC progenitors often eventually occupy the entire epithelial sheet. RNA interference knockdown experiments showed that Dally and Dlp play both partially redundant and distinct roles in regulating Jak/Stat, Wg, and Hh signaling in FSCs. The Drosophila FSC system offers a powerful genetic model to study the mechanisms by which HSPGs exert specific functions in stem cell replacement and competition.
Collapse
|
34
|
Kamath AD, Deehan MA, Frydman HM. Polar cell fate stimulates Wolbachia intracellular growth. Development 2018; 145:dev.158097. [PMID: 29467241 DOI: 10.1242/dev.158097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/12/2018] [Indexed: 11/20/2022]
Abstract
Bacteria are crucial partners in the development and evolution of vertebrates and invertebrates. A large fraction of insects harbor Wolbachia, bacterial endosymbionts that manipulate host reproduction to favor their spreading. Because they are maternally inherited, Wolbachia are under selective pressure to reach the female germline and infect the offspring. However, Wolbachia infection is not limited to the germline. Somatic cell types, including stem cell niches, have higher Wolbachia loads compared with the surrounding tissue. Here, we show a novel Wolbachia tropism to polar cells (PCs), specialized somatic cells in the Drosophila ovary. During oogenesis, all stages of PC development are easily visualized, facilitating the investigation of the kinetics of Wolbachia intracellular growth. Wolbachia accumulation is triggered by particular events of PC morphogenesis, including differentiation from progenitors and between stages 8 and 9 of oogenesis. Moreover, induction of ectopic PC fate is sufficient to promote Wolbachia accumulation. We found that Wolbachia PC tropism is evolutionarily conserved across most Drosophila species, but not in Culex mosquitos. These findings highlight the coordination of endosymbiont tropism with host development and cell differentiation.
Collapse
Affiliation(s)
- Ajit D Kamath
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Mark A Deehan
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Horacio M Frydman
- Department of Biology, Boston University, Boston, MA 02215, USA .,National Emerging Infectious Disease Laboratory, Boston University, Boston, MA 02118, USA
| |
Collapse
|
35
|
Waghmare I, Page-McCaw A. Wnt Signaling in Stem Cell Maintenance and Differentiation in the Drosophila Germarium. Genes (Basel) 2018; 9:genes9030127. [PMID: 29495453 PMCID: PMC5867848 DOI: 10.3390/genes9030127] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 01/19/2023] Open
Abstract
Wnt signaling is a conserved regulator of stem cell behaviors, and the Drosophila germarium has been an important model tissue for the study of stem cell maintenance, differentiation, and proliferation. Here we review Wnt signaling in the germarium, which houses two distinct types of ovarian stem cells: the anteriorly located germline stem cells (GSCs), which give rise to oocytes; and the mid-posteriorly located follicle stem cells (FSCs), which give rise to the somatic follicle cells that cover a developing oocyte. The maintenance and proliferation of GSCs and FSCs is regulated by the stem cell niches, whereas differentiation of the germline is regulated by the differentiation niche. Four distinct Wnt ligands are localized in the germarium, and we focus review on how these Wnt ligands and Wnt signaling affects maintenance and differentiation of both germline and follicle stem cells in their respective niches.
Collapse
Affiliation(s)
- Indrayani Waghmare
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA.
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA.
| |
Collapse
|
36
|
Wang X, Page-McCaw A. Wnt6 maintains anterior escort cells as an integral component of the germline stem cell niche. Development 2018; 145:dev.158527. [PMID: 29361569 PMCID: PMC5818006 DOI: 10.1242/dev.158527] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 12/22/2017] [Indexed: 12/27/2022]
Abstract
Stem cells reside in a niche, a local environment whose cellular and molecular complexity is still being elucidated. In Drosophila ovaries, germline stem cells depend on cap cells for self-renewing signals and physical attachment. Germline stem cells also contact the anterior escort cells, and here we report that anterior escort cells are absolutely required for germline stem cell maintenance. When escort cells die from impaired Wnt signaling or hid expression, the loss of anterior escort cells causes loss of germline stem cells. Anterior escort cells function as an integral niche component by promoting DE-cadherin anchorage and by transiently expressing the Dpp ligand to promote full-strength BMP signaling in germline stem cells. Anterior escort cells are maintained by Wnt6 ligands produced by cap cells; without Wnt6 signaling, anterior escort cells die leaving vacancies in the niche, leading to loss of germline stem cells. Our data identify anterior escort cells as constituents of the germline stem cell niche, maintained by a cap cell-produced Wnt6 survival signal.
Collapse
Affiliation(s)
- Xiaoxi Wang
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| | - Andrea Page-McCaw
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37240, USA
| |
Collapse
|
37
|
Grmai L, Hudry B, Miguel-Aliaga I, Bach EA. Chinmo prevents transformer alternative splicing to maintain male sex identity. PLoS Genet 2018; 14:e1007203. [PMID: 29389999 PMCID: PMC5811060 DOI: 10.1371/journal.pgen.1007203] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 02/13/2018] [Accepted: 01/16/2018] [Indexed: 01/15/2023] Open
Abstract
Reproduction in sexually dimorphic animals relies on successful gamete production, executed by the germline and aided by somatic support cells. Somatic sex identity in Drosophila is instructed by sex-specific isoforms of the DMRT1 ortholog Doublesex (Dsx). Female-specific expression of Sex-lethal (Sxl) causes alternative splicing of transformer (tra) to the female isoform traF. In turn, TraF alternatively splices dsx to the female isoform dsxF. Loss of the transcriptional repressor Chinmo in male somatic stem cells (CySCs) of the testis causes them to "feminize", resembling female somatic stem cells in the ovary. This somatic sex transformation causes a collapse of germline differentiation and male infertility. We demonstrate this feminization occurs by transcriptional and post-transcriptional regulation of traF. We find that chinmo-deficient CySCs upregulate tra mRNA as well as transcripts encoding tra-splice factors Virilizer (Vir) and Female lethal (2)d (Fl(2)d). traF splicing in chinmo-deficient CySCs leads to the production of DsxF at the expense of the male isoform DsxM, and both TraF and DsxF are required for CySC sex transformation. Surprisingly, CySC feminization upon loss of chinmo does not require Sxl but does require Vir and Fl(2)d. Consistent with this, we show that both Vir and Fl(2)d are required for tra alternative splicing in the female somatic gonad. Our work reveals the need for transcriptional regulation of tra in adult male stem cells and highlights a previously unobserved Sxl-independent mechanism of traF production in vivo. In sum, transcriptional control of the sex determination hierarchy by Chinmo is critical for sex maintenance in sexually dimorphic tissues and is vital in the preservation of fertility.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
| | - Bruno Hudry
- MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| | - Irene Miguel-Aliaga
- MRC Clinical Sciences Centre, Imperial College London, London, United Kingdom
| | - Erika A. Bach
- Department of Biochemistry & Molecular Pharmacology, New York University School of Medicine, New York, New York, United States of America
- Kimmel Stem Cell Center, New York University School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
38
|
Su YH, Rastegri E, Kao SH, Lai CM, Lin KY, Liao HY, Wang MH, Hsu HJ. Diet regulates membrane extension and survival of niche escort cells for germline homeostasis via insulin signaling. Development 2018; 145:dev.159186. [DOI: 10.1242/dev.159186] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Accepted: 03/09/2018] [Indexed: 12/29/2022]
Abstract
Diet is an important regulator of stem cell homeostasis, however, the underlying mechanisms of this regulation are not fully known. Here, we report that insulin signaling mediates dietary maintenance of Drosophila ovarian germline stem cells (GSCs) by promoting the extension of niche escort cell (EC) membranes to wrap around GSCs. This wrapping may facilitate the delivery of BMP stemness factors from ECs in the niche to GSCs. In addition to the effects on GSCs, insulin signaling-mediated regulation of EC number and protrusions controls the division and growth of GSC progeny. The effects of insulin signaling on EC membrane extension are, at least in part, driven by enhanced translation of Failed axon connections (Fax) via Ribosomal protein S6 kinase. Fax is a membrane protein that may participate in Abl-regulated cytoskeletal dynamics and is known to be involved in axon bundle formation. Therefore, we conclude that dietary cues stimulate insulin signaling in the niche to regulate EC cellular structure, probably via Fax-dependent cytoskeleton remodeling. This mechanism enhances intercellular contact and facilitates homeostatic interactions between somatic and germline cells in response to diet.
Collapse
Affiliation(s)
- Yu-Han Su
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Elham Rastegri
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei 11529, Taiwan
| | - Shih-Han Kao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Chun-Min Lai
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica and National Chung-Hsing University, Taipei, 11529, Taiwan
- Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung 40227, Taiwan
- Biotechnology Center, National Chung-Hsing University, Taichung 40227, Taiwan
| | - Kun-Yang Lin
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, Academia Sinica and National Chung-Hsing University, Taipei, 11529, Taiwan
- Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung 40227, Taiwan
- Biotechnology Center, National Chung-Hsing University, Taichung 40227, Taiwan
| | - Hung-Yu Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Mu-Hsiang Wang
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Hwei-Jan Hsu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei 11529, Taiwan
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei 11529, Taiwan
- Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
39
|
Dai W, Peterson A, Kenney T, Burrous H, Montell DJ. Quantitative microscopy of the Drosophila ovary shows multiple niche signals specify progenitor cell fate. Nat Commun 2017; 8:1244. [PMID: 29093440 PMCID: PMC5665863 DOI: 10.1038/s41467-017-01322-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 09/09/2017] [Indexed: 01/28/2023] Open
Abstract
Adult stem cells commonly give rise to transit-amplifying progenitors, whose progeny differentiate into distinct cell types. It is unclear if stem cell niche signals coordinate fate decisions within the progenitor pool. Here we use quantitative analysis of Wnt, Hh, and Notch signalling reporters and the cell fate markers Eyes Absent (Eya) and Castor (Cas) to study the effects of hyper-activation and loss of niche signals on progenitor development in the Drosophila ovary. Follicle stem cell (FSC) progeny adopt distinct polar, stalk, and main body cell fates. We show that Wnt signalling transiently inhibits expression of the main body cell fate determinant Eya, and Wnt hyperactivity strongly biases cells towards polar and stalk fates. Hh signalling independently controls the proliferation to differentiation transition. Notch is permissive but not instructive for differentiation of multiple cell types. These findings reveal that multiple niche signals coordinate cell fates and differentiation of progenitor cells.
Collapse
Affiliation(s)
- Wei Dai
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA
| | - Amy Peterson
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA
| | - Thomas Kenney
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA
| | - Haley Burrous
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA
| | - Denise J Montell
- MCDB Department, University of California, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
40
|
The Hippo pathway acts downstream of the Hedgehog signaling to regulate follicle stem cell maintenance in the Drosophila ovary. Sci Rep 2017; 7:4480. [PMID: 28667262 PMCID: PMC5493701 DOI: 10.1038/s41598-017-04052-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 05/09/2017] [Indexed: 11/18/2022] Open
Abstract
The Hippo pathway is conserved and plays important roles in organ size control. The core components of the Hippo pathway are two kinases Hippo (Hpo), Warts (Wts), and a transcription-co-activator Yorkie (Yki). Yki activity is regulated by phosphorylation, which affects its nuclear localization and stability. To determine the role of the Hippo pathway in stem cells, we examine follicle stem cells (FSCs) in the Drosophila ovary. Yki is detected in the nucleus of FSCs. Knockdown of yki in the follicle cell lineage leads to a disruption of the follicular epithelium. Mitotic clones of FSCs mutant for hpo or wts are maintained in the niche and tend to replace the other FSCs, and FSCs mutant for yki are rapidly lost, demonstrating that the Hippo pathway is both required and sufficient for FSC maintenance. Using genetic interaction analyses, we demonstrate that the Hedgehog pathway acts upstream of the Hippo pathway in regulating FSC maintenance. The nuclear localization of Yki is enhanced when the Hedgehog signaling is activated. Furthermore, a constitutively active but not a wild-type Yki promotes FSC maintenance as activation of the Hedgehog signaling does, suggesting that the Hedgehog pathway regulates Yki through a post-translational mechanism in maintaining FSCs.
Collapse
|
41
|
Huang J, Reilein A, Kalderon D. Yorkie and Hedgehog independently restrict BMP production in escort cells to permit germline differentiation in the Drosophila ovary. Development 2017; 144:2584-2594. [PMID: 28619819 DOI: 10.1242/dev.147702] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 06/08/2017] [Indexed: 12/13/2022]
Abstract
Multiple signaling pathways guide the behavior and differentiation of both germline stem cells (GSCs) and somatic follicle stem cells (FSCs) in the Drosophila germarium, necessitating careful control of signal generation, range and responses. Signal integration involves escort cells (ECs), which promote differentiation of the GSC derivatives they envelop, provide niche signals for FSCs and derive directly from FSCs in adults. Hedgehog (Hh) signaling induces the Hippo pathway effector Yorkie (Yki) to promote proliferation and maintenance of FSCs, but Hh also signals to ECs, which are quiescent. Here, we show that in ECs both Hh and Yki limit production of BMP ligands to allow germline differentiation. Loss of Yki produced a more severe germarial phenotype than loss of Hh signaling and principally induced a different BMP ligand. Moreover, Yki activity reporters and epistasis tests showed that Yki does not mediate the key actions of Hh signaling in ECs. Thus, both the coupling and output of the Hh and Yki signaling pathways differ between FSCs and ECs despite their proximity and the fact that FSCs give rise directly to ECs.
Collapse
Affiliation(s)
- Jianhua Huang
- State Key Laboratory of Rice Biology, Institute of Insect Sciences, Zhejiang University, Hangzhou, 310058, China.,Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave., New York, NY 10027, USA
| | - Amy Reilein
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave., New York, NY 10027, USA
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave., New York, NY 10027, USA
| |
Collapse
|
42
|
Teuscher M, Ströhlein N, Birkenbach M, Schultheis D, Schoppmeier M. TC003132 is essential for the follicle stem cell lineage in telotrophic Tribolium oogenesis. Front Zool 2017; 14:26. [PMID: 28533810 PMCID: PMC5438533 DOI: 10.1186/s12983-017-0212-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/11/2017] [Indexed: 12/31/2022] Open
Abstract
Background Stem cells are undifferentiated cells with a potential for self-renewal, which are essential to support normal development and homeostasis. To gain insight into the molecular mechanisms underlying adult stem cell biology and organ evolution, we use the telotrophic ovary of the beetle Tribolium. To this end, we participated in a large-scale RNAi screen in the red flour beetle Tribolium, which identified functions in embryonic and postembryonic development for more than half of the Tribolium genes. Results We identified TC003132 as candidate gene for the follicle stem cell linage in telotrophic Tribolium oogenesis. TC003132 belongs to the Casein Kinase 2 substrate family (CK2S), which in humans is associated with the proliferative activity of different cancers. Upon TC003132 RNAi, central pre-follicular cells are lost, which results in termination of oogenesis. Given that also Notch-signalling is required to promote the mitotic activity of central pre-follicular cells, we performed epistasis experiments with Notch and cut. In addition, we identified a putative follicle stem cell population by monitoring the mitotic pattern of wild type and TC003132 depleted follicle cells by EdU incorporations. In TC003132 RNAi these putative FSCs cease the expression of differentiation makers and are eventually lost. Conclusions TC003132 depleted pre-follicular cells neither react to mitosis or endocycle stimulating signals, suggesting that TC003132 provides competence for differentiation cues. This may resemble the situation in C. elegans were CK2 is required to maintain the balance between proliferation and differentiation in the germ line. Since the earliest effect of TC003132 RNAi is characterized by the loss of putative FSCs, we posit that TC003132 crucially contributes to the proliferation or maintenance of follicle stem cells in the telotrophic Tribolium ovary. Electronic supplementary material The online version of this article (doi:10.1186/s12983-017-0212-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Matthias Teuscher
- Department Biology, Developmental Biology Unit, Friedrich-Alexander-University Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Nadi Ströhlein
- Department Biology, Developmental Biology Unit, Friedrich-Alexander-University Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Markus Birkenbach
- Department Biology, Developmental Biology Unit, Friedrich-Alexander-University Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| | - Dorothea Schultheis
- Present address: Institute of Neuropathology, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Michael Schoppmeier
- Department Biology, Developmental Biology Unit, Friedrich-Alexander-University Erlangen-Nuremberg, Staudtstr. 5, 91058 Erlangen, Germany
| |
Collapse
|
43
|
Reilein A, Melamed D, Park KS, Berg A, Cimetta E, Tandon N, Vunjak-Novakovic G, Finkelstein S, Kalderon D. Alternative direct stem cell derivatives defined by stem cell location and graded Wnt signalling. Nat Cell Biol 2017; 19:433-444. [PMID: 28414313 PMCID: PMC5672635 DOI: 10.1038/ncb3505] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 03/06/2017] [Indexed: 12/15/2022]
Abstract
Adult stem cells provide a renewable source of differentiated cells for a wide variety of tissues and generally give rise to multiple cell types. Basic principles of stem cell organization and regulation underlying this behavior are emerging. Local niche signals maintain stem cells, while different sets of signals act outside the niche to diversify initially equivalent stem cell progeny. Here we show that Drosophila ovarian Follicle Stem Cells (FSCs) produced two distinct cell types directly. This cell fate choice was determined by the A/P position of an FSC and by the magnitude of spatially graded Wnt pathway activity. These findings reveal a paradigm of immediate diversification of stem cell derivatives according to stem cell position within a larger population, guided by a graded niche signal. We also found that FSCs strongly resemble mammalian intestinal stem cells in many aspects of their organization, including population asymmetry and dynamic heterogeneity.
Collapse
Affiliation(s)
- Amy Reilein
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - David Melamed
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Karen Sophia Park
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Ari Berg
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Elisa Cimetta
- Department of Biomedical Engineering, Columbia University, New York, New York 10032, USA
| | - Nina Tandon
- Department of Biomedical Engineering, Columbia University, New York, New York 10032, USA
| | | | - Sarah Finkelstein
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| | - Daniel Kalderon
- Department of Biological Sciences, Columbia University, New York, New York 10027, USA
| |
Collapse
|
44
|
Blake AJ, Finger DS, Hardy VL, Ables ET. RNAi-Based Techniques for the Analysis of Gene Function in Drosophila Germline Stem Cells. Methods Mol Biol 2017; 1622:161-184. [PMID: 28674809 DOI: 10.1007/978-1-4939-7108-4_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Elucidating the full repertoire of molecular mechanisms that promote stem cell maintenance requires sophisticated techniques for identifying and characterizing gene function in stem cells in their native environment. Ovarian germline stem cells in the fruit fly, Drosophila melanogaster, are an ideal model to study the complex molecular mechanisms driving stem cell function in vivo. A variety of new genetic tools make RNAi a useful complement to traditional genetic mutants for the investigation of the molecular mechanisms guiding ovarian germline stem cell function. Here, we provide a detailed guide for using targeted RNAi knockdown for the discovery of gene function in ovarian germline stem cells and their progeny.
Collapse
Affiliation(s)
- Amelia J Blake
- East Carolina University, 1001 E. 10th Street, Mailstop 551, Greenville, NC, 27858, USA
| | - Danielle S Finger
- East Carolina University, 1001 E. 10th Street, Mailstop 551, Greenville, NC, 27858, USA
| | - Victoria L Hardy
- East Carolina University, 1001 E. 10th Street, Mailstop 551, Greenville, NC, 27858, USA
| | - Elizabeth T Ables
- East Carolina University, 1001 E. 10th Street, Mailstop 551, Greenville, NC, 27858, USA.
| |
Collapse
|
45
|
Riechmann V. In vivo RNAi in the Drosophila Follicular Epithelium: Analysis of Stem Cell Maintenance, Proliferation, and Differentiation. Methods Mol Biol 2017; 1622:185-206. [PMID: 28674810 DOI: 10.1007/978-1-4939-7108-4_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In vivo RNAi in Drosophila facilitates simple and rapid analysis of gene functions in a cell- or tissue-specific manner. The versatility of the UAS-GAL4 system allows to control exactly where and when during development the function of a gene is depleted. The epithelium of the ovary is a particularly good model to study in a living animal how stem cells are maintained and how their descendants proliferate and differentiate. Here I provide basic information about the publicly available reagents for in vivo RNAi, and I describe how the oogenesis system can be applied to analyze stem cells and epithelial development at a histological level. Moreover, I give helpful hints to optimize the use of the UAS-GAL4 system for RNAi induction in the follicular epithelium. Finally, I provide detailed step-by-step protocols for ovary dissection, antibody stainings, and ovary mounting for microscopic analysis.
Collapse
Affiliation(s)
- Veit Riechmann
- Medical Faculty Mannheim, Department of Cell and Molecular Biology, Heidelberg University, Ludolf-Krehl-Strasse 13-17, D-68167, Mannheim, Germany.
| |
Collapse
|
46
|
Johnston MJ, Bar-Cohen S, Paroush Z, Nystul TG. Phosphorylated Groucho delays differentiation in the follicle stem cell lineage by providing a molecular memory of EGFR signaling in the niche. Development 2016; 143:4631-4642. [PMID: 27836963 PMCID: PMC5201033 DOI: 10.1242/dev.143263] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 10/31/2016] [Indexed: 01/03/2023]
Abstract
In the epithelial follicle stem cells (FSCs) of the Drosophila ovary, Epidermal Growth Factor Receptor (EGFR) signaling promotes self-renewal, whereas Notch signaling promotes differentiation of the prefollicle cell (pFC) daughters. We have identified two proteins, Six4 and Groucho (Gro), that link the activity of these two pathways to regulate the earliest cell fate decision in the FSC lineage. Our data indicate that Six4 and Gro promote differentiation towards the polar cell fate by promoting Notch pathway activity. This activity of Gro is antagonized by EGFR signaling, which inhibits Gro-dependent repression via p-ERK mediated phosphorylation. We have found that the phosphorylated form of Gro persists in newly formed pFCs, which may delay differentiation and provide these cells with a temporary memory of the EGFR signal. Collectively, these findings demonstrate that phosphorylated Gro labels a transition state in the FSC lineage and describe the interplay between Notch and EGFR signaling that governs the differentiation processes during this period.
Collapse
Affiliation(s)
- Michael J Johnston
- The University of California, San Francisco, Departments of Anatomy and OB-GYN/RS, CA 94122, USA
| | - Shaked Bar-Cohen
- The Hebrew University, Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, Jerusalem 9112102, Israel
| | - Ze'ev Paroush
- The Hebrew University, Department of Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, Jerusalem 9112102, Israel
| | - Todd G Nystul
- The University of California, San Francisco, Departments of Anatomy and OB-GYN/RS, CA 94122, USA
| |
Collapse
|
47
|
Chen D, Chen M, Lu Z, Yang M, Xie L, Zhang W, Xu H, Lu K, Lu Y. Cholesterol induces proliferation of chicken primordial germ cells. Anim Reprod Sci 2016; 171:36-40. [DOI: 10.1016/j.anireprosci.2016.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/18/2016] [Accepted: 05/20/2016] [Indexed: 11/15/2022]
|
48
|
Mottier-Pavie VI, Palacios V, Eliazer S, Scoggin S, Buszczak M. The Wnt pathway limits BMP signaling outside of the germline stem cell niche in Drosophila ovaries. Dev Biol 2016; 417:50-62. [PMID: 27364467 DOI: 10.1016/j.ydbio.2016.06.038] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 04/14/2016] [Accepted: 06/26/2016] [Indexed: 01/07/2023]
Abstract
The mechanisms that modulate and limit the signaling output of adult stem cell niches remain poorly understood. To gain further insights into how these microenvironments are regulated in vivo, we performed a candidate gene screen designed to identify factors that restrict BMP signal production to the cap cells that comprise the germline stem cell (GSC) niche of Drosophila ovaries. Through these efforts, we found that disruption of Wnt4 and components of the canonical Wnt pathway results in a complex germ cell phenotype marked by an expansion of GSC-like cells, pre-cystoblasts and cystoblasts in young females. This phenotype correlates with an increase of decapentaplegic (dpp) mRNA levels within escort cells and varying levels of BMP responsiveness in the germline. Further genetic experiments show that Wnt4, which exhibits graded expression in somatic cells of germaria, activates the Wnt pathway in posteriorly positioned escort cells. The activation of the Wnt pathway appears to be limited by the BMP pathway itself, as loss of Mad in escort cells results in the expansion of Wnt pathway activation. Wnt pathway activity changes within germaria during the course of aging, coincident with changes in dpp production. These data suggest that mutual antagonism between the BMP and Wnt pathways in somatic cells helps to regulate germ cell differentiation.
Collapse
Affiliation(s)
- Violaine I Mottier-Pavie
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Victor Palacios
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Susan Eliazer
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Shane Scoggin
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Michael Buszczak
- Department of Molecular Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA.
| |
Collapse
|
49
|
Lorberbaum DS, Ramos AI, Peterson KA, Carpenter BS, Parker DS, De S, Hillers LE, Blake VM, Nishi Y, McFarlane MR, Chiang AC, Kassis JA, Allen BL, McMahon AP, Barolo S. An ancient yet flexible cis-regulatory architecture allows localized Hedgehog tuning by patched/Ptch1. eLife 2016; 5. [PMID: 27146892 PMCID: PMC4887206 DOI: 10.7554/elife.13550] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/03/2016] [Indexed: 12/24/2022] Open
Abstract
The Hedgehog signaling pathway is part of the ancient developmental-evolutionary animal toolkit. Frequently co-opted to pattern new structures, the pathway is conserved among eumetazoans yet flexible and pleiotropic in its effects. The Hedgehog receptor, Patched, is transcriptionally activated by Hedgehog, providing essential negative feedback in all tissues. Our locus-wide dissections of the cis-regulatory landscapes of fly patched and mouse Ptch1 reveal abundant, diverse enhancers with stage- and tissue-specific expression patterns. The seemingly simple, constitutive Hedgehog response of patched/Ptch1 is driven by a complex regulatory architecture, with batteries of context-specific enhancers engaged in promoter-specific interactions to tune signaling individually in each tissue, without disturbing patterning elsewhere. This structure—one of the oldest cis-regulatory features discovered in animal genomes—explains how patched/Ptch1 can drive dramatic adaptations in animal morphology while maintaining its essential core function. It may also suggest a general model for the evolutionary flexibility of conserved regulators and pathways. DOI:http://dx.doi.org/10.7554/eLife.13550.001
Collapse
Affiliation(s)
- David S Lorberbaum
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States.,Program in Cellular and Molecular Biology, University Of Michigan Medical School, Ann Arbor, United States
| | - Andrea I Ramos
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States.,Program in Cellular and Molecular Biology, University Of Michigan Medical School, Ann Arbor, United States
| | - Kevin A Peterson
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States.,The Jackson Laboratory, Bar Harbor, United States
| | - Brandon S Carpenter
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - David S Parker
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Sandip De
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Lauren E Hillers
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Victoria M Blake
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States.,Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Yuichi Nishi
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California Keck School of Medicine, Los Angeles, United States
| | - Matthew R McFarlane
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States
| | - Ason Cy Chiang
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Judith A Kassis
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United States
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Andrew P McMahon
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, United States.,Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California Keck School of Medicine, Los Angeles, United States
| | - Scott Barolo
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
50
|
Liu Z, Zhong G, Chai PC, Luo L, Liu S, Yang Y, Baeg GH, Cai Y. Coordinated niche-associated signals promote germline homeostasis in the Drosophila ovary. J Cell Biol 2016; 211:469-84. [PMID: 26504174 PMCID: PMC4621830 DOI: 10.1083/jcb.201503033] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Stem cell niches provide localized signaling molecules to promote stem cell fate and to suppress differentiation. The Drosophila melanogaster ovarian niche is established by several types of stromal cells, including terminal filament cells, cap cells, and escort cells (ECs). Here, we show that, in addition to its well-known function as a niche factor expressed in cap cells, the Drosophila transforming growth factor β molecule Decapentaplegic (Dpp) is expressed at a low level in ECs to maintain a pool of partially differentiated germline cells that may dedifferentiate to replenish germline stem cells upon their depletion under normal and stress conditions. Our study further reveals that the Dpp level in ECs is modulated by Hedgehog (Hh) ligands, which originate from both cap cells and ECs. We also demonstrate that Hh signaling exerts its function by suppressing Janus kinase/signal transducer activity, which promotes Dpp expression in ECs. Collectively, our data suggest a complex interplay of niche-associated signals that controls the development of a stem cell lineage.
Collapse
Affiliation(s)
- Zhong Liu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604
| | - Guohua Zhong
- Laboratory of Insect Toxicology, South China Agriculture University, Guangzhou, PR China 510642
| | - Phing Chian Chai
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604
| | - Lichao Luo
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Sen Liu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604
| | - Ying Yang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117543
| | - Gyeong-Hun Baeg
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Yu Cai
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117543
| |
Collapse
|