1
|
Zhao T, Yu XQ. Signaling pathways in Drosophila testis niche: Local signals that regulate stem cell fate. INSECT SCIENCE 2025. [PMID: 40394950 DOI: 10.1111/1744-7917.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/02/2025] [Accepted: 04/16/2025] [Indexed: 05/22/2025]
Abstract
Stem cells are located in a well-structured and specialized microenvironment called the niche. The niche provides signaling molecules to control the survival, self-renewal, and differentiation of stem cells. As tissues generally contain different types of stem cells, it is important to understand how these stem cells are coordinately regulated by various signaling pathways. The Drosophila testis niche serves as an excellent model for studying such processes, because it harbors 2 types of stem cells, germline stem cells and somatic cyst stem cells. In this review, we summarize the roles of key signaling pathways in stem cell maintenance and differentiation in the Drosophila testis.
Collapse
Affiliation(s)
- Ting Zhao
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| | - Xiao-Qiang Yu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou, China
| |
Collapse
|
2
|
Zhang Q, Chen H, Li Z, Qiao J, Liu P, Zheng C, Deng Z, Li X, Zhang H. Bdyof is a Y-chromosome-specific gene required for male development in Bactrocera dorsalis. PEST MANAGEMENT SCIENCE 2025; 81:1785-1793. [PMID: 39611441 DOI: 10.1002/ps.8577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND In many organisms, the Y chromosome contains important genes associated with sex determination and male reproductive development. However, there have been few studies of Y-chromosome-specific genes in non-model species due to the incomplete information of Y chromosome genome and difficulty in sequencing. Here, we screened 90 candidate Y-specific sequences in a constructed transcriptome assembly library by using the chromosome quotient method, among which 11 were unreported sequences associated with male reproductive development, including Bactrocera dorsalis Y-specific Oligozoospermia factor (Bdyof) with the highest expression in the testis. RESULTS CRISPR/Cas9-mediated knockout of Bdyof resulted in abnormal testis development, significantly reduced sperm count, and obviously lower egg hatching rate in homozygous mutant flies. In addition, Bdyof knockout decreased the expression of dsx-M. CONCLUSION This results provides new insights into the biological processes related to male reproductive development controlled by the Y-chromosome-specific gene Bdyof, thus providing a promising molecular target for the study of agricultural pests. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Qiuyuan Zhang
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hao Chen
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ziniu Li
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Jiao Qiao
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Peipei Liu
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Chenjun Zheng
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhurong Deng
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaoxue Li
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hongyu Zhang
- National Key Laboratory for Germplasm Innovation and Utilization for Fruit and Vegetable Horticultural Crops, Hubei Hongshan Laboratory, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
3
|
Nelson KA, Lenhart KF, Anllo L, DiNardo S. The Drosophila hematopoietic niche assembles through collective cell migration controlled by neighbor tissues and Slit-Robo signaling. eLife 2025; 13:RP100455. [PMID: 39750120 PMCID: PMC11698496 DOI: 10.7554/elife.100455] [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] [Indexed: 01/04/2025] Open
Abstract
Niches are often found in specific positions in tissues relative to the stem cells they support. Consistency of niche position suggests that placement is important for niche function. However, the complexity of most niches has precluded a thorough understanding of how their proper placement is established. To address this, we investigated the formation of a genetically tractable niche, the Drosophila Posterior Signaling Center (PSC), the assembly of which had not been previously explored. This niche controls hematopoietic progenitors of the lymph gland (LG). PSC cells were previously shown to be specified laterally in the embryo, but ultimately reside dorsally, at the LG posterior. Here, using live-imaging, we show that PSC cells migrate as a tight collective and associate with multiple tissues during their trajectory to the LG posterior. We find that Slit emanating from two extrinsic sources, visceral mesoderm and cardioblasts, is required for the PSC to remain a collective, and for its attachment to cardioblasts during migration. Without proper Slit-Robo signaling, PSC cells disperse, form aberrant contacts, and ultimately fail to reach their stereotypical position near progenitors. Our work characterizes a novel example of niche formation and identifies an extrinsic signaling relay that controls precise niche positioning.
Collapse
Affiliation(s)
- Kara A Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Kari F Lenhart
- Department of Biology, Drexel UniversityPhiladelphiaUnited States
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaUnited States
- Institute for Regenerative Medicine at the University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
4
|
Vida GS, Botto E, DiNardo S. Maintenance of niche architecture requires actomyosin and enables proper stem cell signaling and oriented division in the Drosophila testis. Development 2025; 152:dev204498. [PMID: 39620974 PMCID: PMC11795290 DOI: 10.1242/dev.204498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/20/2024] [Indexed: 12/13/2024]
Abstract
Stem cells are essential to repair and regenerate tissues, and often reside in a niche that controls their behavior. Here, we use the Drosophila testis niche, a paradigm for niche-stem cell interactions, to address the cell biological features that maintain niche structure and function during its steady-state operation. We report enrichment of Myosin II (MyoII) and a key regulator of actomyosin contractility (AMC), Rho Kinase (ROK), within the niche cell cortex at the interface with germline stem cells (GSCs). Compromising MyoII and ROK disrupts niche architecture, suggesting that AMC in niche cells is important to maintain its reproducible structure. Furthermore, defects in niche architecture disrupt GSC function. Our data suggest that the niche signals less robustly to adjacent germ cells yet permits increased numbers of cells to respond to the signal. Finally, compromising MyoII in niche cells leads to increased misorientation of centrosomes in GSCs as well as defects in the centrosome orientation checkpoint. Ultimately, this work identifies a crucial role for AMC-dependent maintenance of niche structure to ensure a proper complement of stem cells that correctly execute divisions.
Collapse
Affiliation(s)
- Gabriela S. Vida
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA
- The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | - Elizabeth Botto
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA
- The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, USA
- The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Grmai L, Mychalczuk M, Arkalgud A, Vasudevan D. Sexually dimorphic ATF4 expression in the fat confers female stress tolerance in Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630478. [PMID: 39763862 PMCID: PMC11703189 DOI: 10.1101/2024.12.27.630478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Metabolic differences between males and females have been well documented across many species. However, the molecular basis of these differences and how they impact tolerance to nutrient deprivation is still under investigation. In this work, we use Drosophila melanogaster to demonstrate that sex-specific differences in fat tissue metabolism are driven, in part, by dimorphic expression of the Integrated Stress Response (ISR) transcription factor, ATF4. We found that female fat tissues have higher ATF4 activity than their male counter parts under homeostatic conditions. This dimorphism was partly due to a female bias in transcript abundance of specific ATF4 splice isoforms. We found that the canonical sex determinants transformer (tra) and doublesex (dsx) drive such dimorphic ATF4 transcript abundance. These differences persist in a genetic model of nutrient deprivation, where female animals showed greater resistance to lethality than males in an ATF4-dependent manner. These results suggest that higher ATF4 activity confers higher tolerance to stress in females. Together, our data describe a previously unknown facet of ISR signaling wherein sexual identity of adipose tissue confers differential stress tolerance in males and females. Since energy storage mechanisms are known to be dimorphic and have been linked to ATF4 regulation, our studies provide a mechanistic starting point for understanding how sexual identity influences metabolic disease outcomes.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Present Address: Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Melissa Mychalczuk
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Present Address: Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Aditya Arkalgud
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Deepika Vasudevan
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
6
|
Yang Q, Wijaya F, Kapoor R, Chandrasekaran H, Jagtiani S, Moran I, Hime GR. Unusual modes of cell and nuclear divisions characterise Drosophila development. Biochem Soc Trans 2024; 52:2281-2295. [PMID: 39508395 PMCID: PMC11668308 DOI: 10.1042/bst20231341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
The growth and development of metazoan organisms is dependent upon a co-ordinated programme of cellular proliferation and differentiation, from the initial formation of the zygote through to maintenance of mature organs in adult organisms. Early studies of proliferation of ex vivo cultures and unicellular eukaryotes described a cyclic nature of cell division characterised by periods of DNA synthesis (S-phase) and segregation of newly synthesized chromosomes (M-phase) interspersed by seeming inactivity, the gap phases, G1 and G2. We now know that G1 and G2 play critical roles in regulating the cell cycle, including monitoring of favourable environmental conditions to facilitate cell division, and ensuring genomic integrity prior to DNA replication and nuclear division. M-phase is usually followed by the physical separation of nascent daughters, termed cytokinesis. These phases where G1 leads to S phase, followed by G2 prior to M phase and the subsequent cytokinesis to produce two daughters, both identical in genomic composition and cellular morphology are what might be termed an archetypal cell division. Studies of development of many different organs in different species have demonstrated that this stereotypical cell cycle is often subverted to produce specific developmental outcomes, and examples from over 100 years of analysis of the development of Drosophila melanogaster have uncovered many different modes of cell division within this one species.
Collapse
Affiliation(s)
- Qiaolin Yang
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Fernando Wijaya
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Ridam Kapoor
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Harshaa Chandrasekaran
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Siddhant Jagtiani
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Izaac Moran
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gary R. Hime
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
7
|
Zambrano-Tipan D, Narváez-Padilla V, Reynaud E. Escargot a Snail superfamily member and its multiple roles in Drosophila melanogaster development. J Cell Physiol 2024; 239:e31269. [PMID: 38572978 DOI: 10.1002/jcp.31269] [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: 12/01/2023] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024]
Abstract
The Snail superfamily of transcription factors plays a crucial role in metazoan development; one of the most important vertebrate members of this family is Snai1 which is orthologous to the Drosophila melanogaster esg gene. This review offers a comprehensive examination of the roles of the esg gene in Drosophila development, covering its expression pattern and downstream targets, and draws parallels between the vertebrate Snai1 family proteins on controlling the epithelial-to-mesenchymal transition and esg. This gene regulates stemness, ploidy, and pluripontency. esg is expressed in various tissues during development, including the gut, imaginal discs, and neuroblasts. The functions of the esg include the suppression of differentiation in intestinal stem cells and the preservation of diploidy in imaginal cells. In the nervous system development, esg expression also inhibits neuroblast differentiation, thus regulating the number of neurons and the moment in development of neuronal differentiation. Loss of esg function results in diverse developmental defects, including defects in intestinal stem cell maintenance and differentiation, and alters imaginal disc and nervous system development. Expression levels of esg also play a role in regulating longevity and metabolism in adult stages. This review provides an overview of the current understanding of esg's developmental role, emphasizing cellular and tissue effects that arise from its loss of function. The insights gained may contribute to a better understanding of evolutionary conserved developmental mechanisms and certain metabolic diseases.
Collapse
Affiliation(s)
- Diego Zambrano-Tipan
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Verónica Narváez-Padilla
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Enrique Reynaud
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
8
|
Nelson KA, Lenhart KF, Anllo L, DiNardo S. The Drosophila hematopoietic niche assembles through collective cell migration controlled by neighbor tissues and Slit-Robo signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.600069. [PMID: 38979182 PMCID: PMC11230208 DOI: 10.1101/2024.06.21.600069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Niches are often found in specific positions in tissues relative to the stem cells they support. Consistency of niche position suggests that placement is important for niche function. However, the complexity of most niches has precluded a thorough understanding of how their proper placement is established. To address this, we investigated the formation of a genetically tractable niche, the Drosophila Posterior Signaling Center (PSC), the assembly of which had not been previously explored. This niche controls hematopoietic progenitors of the lymph gland (LG). PSC cells were previously shown to be specified laterally in the embryo, but ultimately reside dorsally, at the LG posterior. Here, using live-imaging, we show that PSC cells migrate as a tight collective and associate with multiple tissues during their trajectory to the LG posterior. We find that Slit emanating from two extrinsic sources, visceral mesoderm and cardioblasts, is required for the PSC to remain a collective, and for its attachment to cardioblasts during migration. Without proper Slit-Robo signaling, PSC cells disperse, form aberrant contacts, and ultimately fail to reach their stereotypical position near progenitors. Our work characterizes a novel example of niche formation and identifies an extrinsic signaling relay that controls precise niche positioning.
Collapse
Affiliation(s)
- Kara A Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
| | - Kari F Lenhart
- Department of Biology, Drexel University, 3245 Chestnut St. Philadelphia, PA 19104, United States
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
- Present address: Department of Biology, East Carolina University, 458 Science & Tech Bldg. Greenville, NC 27858, United States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd. Philadelphia, PA 19104, United States
- Institute for Regenerative Medicine at the University of Pennsylvania, 3400 Civic Center Blvd. Philadelphia, PA 19104, United States
| |
Collapse
|
9
|
Kong R, Zhao H, Li J, Ma Y, Li N, Shi L, Li Z. A regulatory loop of JAK/STAT signalling and its downstream targets represses cell fate conversion and maintains male germline stem cell niche homeostasis. Cell Prolif 2024; 57:e13648. [PMID: 38987866 PMCID: PMC11471429 DOI: 10.1111/cpr.13648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/03/2024] [Accepted: 04/11/2024] [Indexed: 07/12/2024] Open
Abstract
A specialised microenvironment, termed niche, provides extrinsic signals for the maintenance of residential stem cells. However, how residential stem cells maintain niche homeostasis and whether stromal niche cells could convert their fate into stem cells to replenish lost stem cells upon systemic stem cell loss remain largely unknown. Here, through systemic identification of JAK/STAT downstream targets in adult Drosophila testis, we show that Escargot (Esg), a member of the Snail family of transcriptional factors, is a putative JAK/STAT downstream target. esg is intrinsically required in cyst stem cells (CySCs) but not in germline stem cells (GSCs). esg depletion in CySCs results in CySC loss due to differentiation and non-cell autonomous GSC loss. Interestingly, hub cells are gradually lost by delaminating from the hub and converting into CySCs in esg-defective testes. Mechanistically, esg directly represses the expression of socs36E, the well-known downstream target and negative regulator of JAK/STAT signalling. Finally, further depletion of socs36E completely rescues the defects observed in esg-defective testes. Collectively, JAK/STAT target Esg suppresses SOCS36E to maintain CySC fate and repress niche cell conversion. Thus, our work uncovers a regulatory loop between JAK/STAT signalling and its downstream targets in controlling testicular niche homeostasis under physiological conditions.
Collapse
Affiliation(s)
- Ruiyan Kong
- Laboratory of Stem Cell Biology, College of Life SciencesCapital Normal UniversityBeijingChina
| | - Hang Zhao
- Laboratory of Stem Cell Biology, College of Life SciencesCapital Normal UniversityBeijingChina
| | - Juan Li
- Laboratory of Stem Cell Biology, College of Life SciencesCapital Normal UniversityBeijingChina
| | - Yankun Ma
- Laboratory of Stem Cell Biology, College of Life SciencesCapital Normal UniversityBeijingChina
| | - Ningfang Li
- Laboratory of Stem Cell Biology, College of Life SciencesCapital Normal UniversityBeijingChina
| | - Lin Shi
- Laboratory of Stem Cell Biology, College of Life SciencesCapital Normal UniversityBeijingChina
| | - Zhouhua Li
- Laboratory of Stem Cell Biology, College of Life SciencesCapital Normal UniversityBeijingChina
| |
Collapse
|
10
|
Warder BN, Nelson KA, Sui J, Anllo L, DiNardo S. An actomyosin network organizes niche morphology and responds to feedback from recruited stem cells. Curr Biol 2024; 34:3917-3930.e6. [PMID: 39137785 PMCID: PMC11387155 DOI: 10.1016/j.cub.2024.07.041] [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: 09/28/2023] [Revised: 04/18/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024]
Abstract
Stem cells often rely on signals from a niche, which in many tissues adopts a precise morphology. What remains elusive is how niches are formed and how morphology impacts function. To address this, we leverage the Drosophila gonadal niche, which affords genetic tractability and live-imaging. We have previously shown mechanisms dictating niche cell migration to their appropriate position within the gonad and the resultant consequences on niche function. Here, we show that once positioned, niche cells robustly polarize filamentous actin (F-actin) and non-muscle myosin II (MyoII) toward neighboring germ cells. Actomyosin tension along the niche periphery generates a highly reproducible smoothened contour. Without contractility, niches are misshapen and exhibit defects in their ability to regulate germline stem cell behavior. We additionally show that germ cells aid in polarizing MyoII within niche cells and that extrinsic input is required for niche morphogenesis and function. Our work reveals a feedback mechanism where stem cells shape the niche that guides their behavior.
Collapse
Affiliation(s)
- Bailey N Warder
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kara A Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin Sui
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Warder BN, Nelson KA, Sui J, Anllo L, DiNardo S. An actomyosin network organizes niche morphology and responds to feedback from recruited stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.08.556877. [PMID: 38746236 PMCID: PMC11092431 DOI: 10.1101/2023.09.08.556877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Stem cells often rely on signals from a niche, which in many tissues adopts a precise morphology. What remains elusive is how niches are formed, and how morphology impacts function. To address this, we leverage the Drosophila gonadal niche, which affords genetic tractability and live-imaging. We have previously shown mechanisms dictating niche cell migration to their appropriate position within the gonad, and the resultant consequences on niche function. Here, we show that once positioned, niche cells robustly polarize filamentous actin (F-actin) and Non-muscle Myosin II (MyoII) towards neighboring germ cells. Actomyosin tension along the niche periphery generates a highly reproducible smoothened contour. Without contractility, niches are misshapen and exhibit defects in their ability to regulate germline stem cell behavior. We additionally show that germ cells aid in polarizing MyoII within niche cells, and that extrinsic input is required for niche morphogenesis and function. Our work reveals a feedback mechanism where stem cells shape the niche that guides their behavior.
Collapse
Affiliation(s)
- Bailey N. Warder
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kara A. Nelson
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Justin Sui
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Vidaurre V, Song A, Li T, Ku WL, Zhao K, Qian J, Chen X. The Drosophila histone methyltransferase SET1 coordinates multiple signaling pathways in regulating male germline stem cell maintenance and differentiation. Development 2024; 151:dev202729. [PMID: 39007366 PMCID: PMC11369688 DOI: 10.1242/dev.202729] [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: 01/25/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Many tissue-specific adult stem cell lineages maintain a balance between proliferation and differentiation. Here, we study how the H3K4me3 methyltransferase Set1 regulates early-stage male germ cells in Drosophila. Early-stage germline-specific knockdown of Set1 results in temporally progressive defects, arising as germ cell loss and developing into overpopulated early-stage germ cells. These germline defects also impact the niche architecture and cyst stem cell lineage non-cell-autonomously. Additionally, wild-type Set1, but not the catalytically inactive Set1, rescues the Set1 knockdown phenotypes, highlighting the functional importance of the methyltransferase activity of Set1. Further, RNA-sequencing experiments reveal key signaling pathway components, such as the JAK-STAT pathway gene Stat92E and the BMP pathway gene Mad, which are upregulated upon Set1 knockdown. Genetic interaction assays support the functional relationships between Set1 and JAK-STAT or BMP pathways, as both Stat92E and Mad mutations suppress the Set1 knockdown phenotypes. These findings enhance our understanding of the balance between proliferation and differentiation in an adult stem cell lineage. The phenotype of germ cell loss followed by over-proliferation when inhibiting a histone methyltransferase also raises concerns about using their inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Velinda Vidaurre
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Annabelle Song
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Taibo Li
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wai Lim Ku
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20814, USA
| | - Keji Zhao
- Laboratory of Epigenome Biology, Systems Biology Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20814, USA
| | - Jiang Qian
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xin Chen
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, 4000 Jones Bridge Road, Chevy Chase, MD 20815, USA
| |
Collapse
|
13
|
Grmai L, Jimenez E, Baxter E, Doren MV. Steroid signaling controls sex-specific development in an invertebrate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573099. [PMID: 38187640 PMCID: PMC10769319 DOI: 10.1101/2023.12.22.573099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
In vertebrate sexual development, two important steroid hormones, testosterone and estrogen, regulate the sex-specific development of many tissues. In contrast, invertebrates utilize a single steroid hormone, ecdysone, to regulate developmental timing in both sexes. However, here we show that in Drosophila melanogaster, sex-specific ecdysone (E) signaling controls important aspects of gonad sexual dimorphism. Rather than being regulated at the level of hormone production, hormone activity is regulated cell-autonomously through sex-specific hormone reception. Ecdysone receptor (EcR) expression is restricted to the developing ovary and is repressed in the testis at a time when ecdysone initiates ovary morphogenesis. Interestingly, EcR expression is regulated downstream of the sex determination factor Doublesex (Dsx), the founding member of the Dsx/Mab3 Related Transcription Factor (DMRT) family that regulates gonad development in all animals. E signaling is required for normal ovary development1,2, and ectopic activation of E signaling in the testis antagonized stem cell niche identity and feminized somatic support cells, which were transformed into follicle-like cells. This work demonstrates that invertebrates can also use steroid hormone signaling to control sex-specific development. Further, it may help explain recent work showing that vertebrate sexual development is surprisingly cell-autonomous. For example, chickens utilize testosterone and estrogen to control sex-specific development, but when they have a mixture of cells with male and female genotypes, the male cells develop as male and the female cells develop as female despite exposure to the same circulating hormones3. Sex-specific regulation of steroid hormone signaling may well underly such cell-autonomous sexual fate choices in vertebrates as it does in Drosophila.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Erin Jimenez
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ellen Baxter
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mark Van Doren
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Chen X, Qi Y, Huang Q, Sun C, Zheng Y, Ji L, Shi Y, Cheng X, Li Z, Zheng S, Cao Y, Gu Z, Yu J. Single-cell transcriptome characteristics of testicular terminal epithelium lineages during aging in the Drosophila. Aging Cell 2024; 23:e14057. [PMID: 38044573 PMCID: PMC10928582 DOI: 10.1111/acel.14057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/05/2023] Open
Abstract
Aging is a complex biological process leading to impaired functions, with a variety of hallmarks. In the testis of Drosophila, the terminal epithelium region is involved in spermatid release and maturation, while its functional diversity and regulatory mechanism remain poorly understood. In this study, we performed single-cell RNA-sequencing analysis (scRNA-seq) to characterize the transcriptomes of terminal epithelium in Drosophila testes at 2-, 10 and 40-Days. Terminal epithelium populations were defined with Metallothionein A (MtnA) and subdivided into six novel sub-cell clusters (EP0-EP5), and a series of marker genes were identified based on their expressions. The data revealed the functional characteristics of terminal epithelium populations, such as tight junction, focal adhesion, bacterial invasion, oxidative stress, mitochondrial function, proteasome, apoptosis and metabolism. Interestingly, we also found that disrupting genes for several relevant pathways in terminal epithelium led to male fertility disorders. Moreover, we also discovered a series of age-biased genes and pseudotime trajectory mediated state-biased genes during terminal epithelium aging. Differentially expressed genes during terminal epithelium aging were mainly participated in the regulation of several common signatures, e.g. mitochondria-related events, protein synthesis and degradation, and metabolic processes. We further explored the Drosophila divergence and selection in the functional constraints of age-biased genes during aging, revealing that age-biased genes in epithelial cells of 2 Days group evolved rapidly and were endowed with greater evolutionary advantages. scRNA-seq analysis revealed the diversity of testicular terminal epithelium populations, providing a gene target resource for further systematic research of their functions during aging.
Collapse
Affiliation(s)
- Xia Chen
- Department of Obstetrics and Gynecology, Nantong First People's HospitalAffiliated Hospital 2 of Nantong University; Medical School of Nantong University, Nantong UniversityNantongJiangsuChina
| | - Yujuan Qi
- Clinical Center of Reproductive Medicine, Xuzhou Central HospitalXuzhou Clinical School of Xuzhou Medical UniversityXuzhouChina
| | - Qiuru Huang
- Institute of Reproductive MedicineMedical School of Nantong University, Nantong UniversityNantongChina
| | - Chi Sun
- Department of GeriatricsAffiliated Hospital of Nantong University, Nantong UniversityNantongChina
| | - Yanli Zheng
- Department of Obstetrics and Gynecology, Nantong First People's HospitalAffiliated Hospital 2 of Nantong University; Medical School of Nantong University, Nantong UniversityNantongJiangsuChina
| | - Li Ji
- Institute of Reproductive MedicineMedical School of Nantong University, Nantong UniversityNantongChina
| | - Yi Shi
- Institute of Reproductive MedicineMedical School of Nantong University, Nantong UniversityNantongChina
| | - Xinmeng Cheng
- Institute of Reproductive MedicineMedical School of Nantong University, Nantong UniversityNantongChina
| | - Zhenbei Li
- Clinical Center of Reproductive Medicine, Xuzhou Central HospitalXuzhou Clinical School of Xuzhou Medical UniversityXuzhouChina
| | - Sen Zheng
- Clinical Center of Reproductive Medicine, Xuzhou Central HospitalXuzhou Clinical School of Xuzhou Medical UniversityXuzhouChina
| | - Yijuan Cao
- Clinical Center of Reproductive Medicine, Xuzhou Central HospitalXuzhou Clinical School of Xuzhou Medical UniversityXuzhouChina
| | - Zhifeng Gu
- Department of RheumatologyAffiliated Hospital of Nantong University, Nantong UniversityNantongChina
| | - Jun Yu
- Institute of Reproductive MedicineMedical School of Nantong University, Nantong UniversityNantongChina
| |
Collapse
|
15
|
Eslahi M, Nematbakhsh N, Dastmalchi N, Teimourian S, Safaralizadeh R. Signaling Pathways in Drosophila gonadal Stem Cells. Curr Stem Cell Res Ther 2024; 19:154-165. [PMID: 36788694 DOI: 10.2174/1574888x18666230213144531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 02/16/2023]
Abstract
The stem cells' ability to divide asymmetrically to produce differentiating and self-renewing daughter cells is crucial to maintain tissue homeostasis and development. Stem cell maintenance and differentiation rely on their regulatory microenvironment termed 'niches'. The mechanisms of the signal transduction pathways initiated from the niche, regulation of stem cell maintenance and differentiation were quite challenging to study. The knowledge gained from the study of Drosophila melanogaster testis and ovary helped develop our understanding of stem cell/niche interactions and signal pathways related to the regulatory mechanisms in maintaining homeostasis of adult tissue. In this review, we discuss the role of signaling pathways in Drosophila gonadal stem cell regeneration, competition, differentiation, dedifferentiation, proliferation, and fate determination. Furthermore, we present the current knowledge on how these signaling pathways are implicated in cancer, and how they contribute as potential candidates for effective cancer treatment.
Collapse
Affiliation(s)
- Maede Eslahi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Negin Nematbakhsh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Narges Dastmalchi
- Department of Biology, University College of Nabi Akram, Tabriz, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
16
|
Sreejith P, Kim C. Lin28 is Required for Single Niche Development in the Drosophila Male Gonad. Dev Reprod 2023; 27:221-226. [PMID: 38292237 PMCID: PMC10824566 DOI: 10.12717/dr.2023.27.4.221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/22/2023] [Accepted: 11/23/2023] [Indexed: 02/01/2024]
Abstract
A stem cell niche provides an environment that governs stem cell maintenance and division. Thus, the development of a proper niche is of prime importance to stem cell behaviors. Mechanisms of niche development are beginning to be revealed in the Drosophila male gonad. Niche cells are initially dispersed throughout the gonad, then assemble at its apical tip through the anterior migration of posteriorly located niche cells. The molecular mechanisms of this migration and assembly are still poorly understood. Here we show evidence suggesting that Lin28, an RNA-binding protein and regulator of let7 genesis, might be an intrinsic factor for the anterior migration of niche cells. We found that a dispersed, ectopic niche, a phenotype observed with anterior migration defects, occurs in lin28 mutant gonads. This phenotype is rescued by expression of lin28 in the niche cells. These findings suggest that Lin28 might be required for the anterior migration of niche cells.
Collapse
Affiliation(s)
- Perinthottathil Sreejith
- Department of Biomedical Genetics,
University of Rochester Medical Center, Rochester,
NY 14642, USA
| | - Changsoo Kim
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| |
Collapse
|
17
|
Nagai H, Nagai LAE, Tasaki S, Nakato R, Umetsu D, Kuranaga E, Miura M, Nakajima Y. Nutrient-driven dedifferentiation of enteroendocrine cells promotes adaptive intestinal growth in Drosophila. Dev Cell 2023; 58:1764-1781.e10. [PMID: 37689060 DOI: 10.1016/j.devcel.2023.08.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 05/05/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Post-developmental organ resizing improves organismal fitness under constantly changing nutrient environments. Although stem cell abundance is a fundamental determinant of adaptive resizing, our understanding of its underlying mechanisms remains primarily limited to the regulation of stem cell division. Here, we demonstrate that nutrient fluctuation induces dedifferentiation in the Drosophila adult midgut to drive adaptive intestinal growth. From lineage tracing and single-cell RNA sequencing, we identify a subpopulation of enteroendocrine (EE) cells that convert into functional intestinal stem cells (ISCs) in response to dietary glucose and amino acids by activating the JAK-STAT pathway. Genetic ablation of EE-derived ISCs severely impairs ISC expansion and midgut growth despite the retention of resident ISCs, and in silico modeling further indicates that EE dedifferentiation enables an efficient increase in the midgut cell number while maintaining epithelial cell composition. Our findings identify a physiologically induced dedifferentiation that ensures ISC expansion during adaptive organ growth in concert with nutrient conditions.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| | | | - Sohei Tasaki
- Graduate School of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Ryuichiro Nakato
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Erina Kuranaga
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuichiro Nakajima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| |
Collapse
|
18
|
Yildirim K, van Nierop Y Sanchez P, Lohmann I. Analysis of Bub3 and Nup75 in the Drosophila male germline lineage. Cells Dev 2023; 175:203863. [PMID: 37286104 DOI: 10.1016/j.cdev.2023.203863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/04/2023] [Accepted: 06/01/2023] [Indexed: 06/09/2023]
Abstract
Extensive communication at the stem cell-niche interface and asymmetric stem cell division is key for the homeostasis of the Drosophila male germline stem cell system. To improve our understanding of these processes, we analysed the function of the mitotic checkpoint complex (MCC) component Bub3 and the nucleoporin Nup75, a component of the nuclear pore complex realizing the transport of signalling effector molecules to the nucleus, in the Drosophila testis. By lineage-specific interference, we found that the two genes control germline development and maintenance. Bub3 is continuously required in the germline, as its loss results in the beginning in an over-proliferation of early germ cells and later on in loss of the germline. The absence of the germline lineage in such testes has dramatic cell non-autonomous consequences, as cells co-expressing markers of hub and somatic cyst cell fates accumulate and populate in extreme cases the whole testis. Our analysis of Nups showed that some of them are critical for lineage maintenance, as their depletion results in the loss of the affected lineage. In contrast, Nup75 plays a role in controlling proliferation of early germ cells but not differentiating spermatogonia and seems to be involved in keeping hub cells quiescent. In sum, our analysis shows that Bub3 and Nup75 are required for male germline development and maintenance.
Collapse
Affiliation(s)
- Kerem Yildirim
- Heidelberg University, Centre for Organismal Studies (COS) Heidelberg, Department of Developmental Biology and Cell Networks - Cluster of Excellence, Heidelberg, Germany
| | - Patrick van Nierop Y Sanchez
- Heidelberg University, Centre for Organismal Studies (COS) Heidelberg, Department of Developmental Biology and Cell Networks - Cluster of Excellence, Heidelberg, Germany
| | - Ingrid Lohmann
- Heidelberg University, Centre for Organismal Studies (COS) Heidelberg, Department of Developmental Biology and Cell Networks - Cluster of Excellence, Heidelberg, Germany.
| |
Collapse
|
19
|
Canal Domenech B, Fricke C. Developmental heat stress interrupts spermatogenesis inducing early male sterility in Drosophila melanogaster. J Therm Biol 2023; 114:103589. [PMID: 37300998 DOI: 10.1016/j.jtherbio.2023.103589] [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: 03/21/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 06/12/2023]
Abstract
Thermal stress leads to fertility reduction, can cause temporal sterility and thus results in fitness loss with severe ecological and evolutionary consequences, e.g., threatening species persistence already at sub-lethal temperatures. For males we here tested which developmental stage is particularly sensitive to heat stress in the model species Drosophila melanogaster. As developmental stages characterize the different steps of sperm development, we could narrow down which particular processes are heat sensitive. We studied early male reproductive ability and, by following recovery dynamics after a move to benign temperatures, we investigated general mechanisms behind a subsequent gain of fertility. We found strong support to suggest that the last steps of spermatogenesis are particularly sensitive to heat stress, as processes occurring during the pupal stage were mostly interrupted, delaying both sperm production and sperm maturation. Moreover, further measurements in the testes and for proxies of sperm availability indicating the onset of adult reproductive capacity matched the expected heat-induced delay in completing spermatogenesis. We discuss these results within the context of how heat stress affects reproductive organ function and the consequences for male reproductive potential.
Collapse
Affiliation(s)
- Berta Canal Domenech
- Institute for Evolution and Biodiversity, University of Muenster, Muenster, Germany; Muenster Graduate School of Evolution, University of Muenster, Muenster, Germany.
| | - Claudia Fricke
- Institute for Evolution and Biodiversity, University of Muenster, Muenster, Germany; Institute for Zoology, Halle-Wittenberg University, Halle (Saale), Germany.
| |
Collapse
|
20
|
Raz AA, Vida GS, Stern SR, Mahadevaraju S, Fingerhut JM, Viveiros JM, Pal S, Grey JR, Grace MR, Berry CW, Li H, Janssens J, Saelens W, Shao Z, Hu C, Yamashita YM, Przytycka T, Oliver B, Brill JA, Krause H, Matunis EL, White-Cooper H, DiNardo S, Fuller MT. Emergent dynamics of adult stem cell lineages from single nucleus and single cell RNA-Seq of Drosophila testes. eLife 2023; 12:e82201. [PMID: 36795469 PMCID: PMC9934865 DOI: 10.7554/elife.82201] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/19/2023] [Indexed: 02/17/2023] Open
Abstract
Proper differentiation of sperm from germline stem cells, essential for production of the next generation, requires dramatic changes in gene expression that drive remodeling of almost all cellular components, from chromatin to organelles to cell shape itself. Here, we provide a single nucleus and single cell RNA-seq resource covering all of spermatogenesis in Drosophila starting from in-depth analysis of adult testis single nucleus RNA-seq (snRNA-seq) data from the Fly Cell Atlas (FCA) study. With over 44,000 nuclei and 6000 cells analyzed, the data provide identification of rare cell types, mapping of intermediate steps in differentiation, and the potential to identify new factors impacting fertility or controlling differentiation of germline and supporting somatic cells. We justify assignment of key germline and somatic cell types using combinations of known markers, in situ hybridization, and analysis of extant protein traps. Comparison of single cell and single nucleus datasets proved particularly revealing of dynamic developmental transitions in germline differentiation. To complement the web-based portals for data analysis hosted by the FCA, we provide datasets compatible with commonly used software such as Seurat and Monocle. The foundation provided here will enable communities studying spermatogenesis to interrogate the datasets to identify candidate genes to test for function in vivo.
Collapse
Affiliation(s)
- Amelie A Raz
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Howard Hughes Medical InstituteCambridgeUnited States
| | - Gabriela S Vida
- Department of Cell and Developmental Biology, The Perelman School of Medicine and The Penn Institute for Regenerative MedicinePhiladelphiaUnited States
| | - Sarah R Stern
- Department of Developmental Biology, Stanford University School of MedicineStanfordUnited States
| | - Sharvani Mahadevaraju
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaUnited States
| | - Jaclyn M Fingerhut
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Howard Hughes Medical InstituteCambridgeUnited States
| | - Jennifer M Viveiros
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Soumitra Pal
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of HealthBethesdaUnited States
| | - Jasmine R Grey
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Mara R Grace
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Cameron W Berry
- Department of Developmental Biology, Stanford University School of MedicineStanfordUnited States
| | - Hongjie Li
- Huffington Center on Aging and Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Jasper Janssens
- JVIB Center for Brain & Disease Research, and the Department of Human Genetics, KU LeuvenLeuvenBelgium
| | - Wouter Saelens
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, and Department of Applied Mathematics, Computer Science and Statistics, Ghent UniversityGhentBelgium
| | - Zhantao Shao
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Chun Hu
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Yukiko M Yamashita
- Whitehead Institute for Biomedical Research and Department of Biology, Massachusetts Institute of Technology, Howard Hughes Medical InstituteCambridgeUnited States
| | - Teresa Przytycka
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of HealthBethesdaUnited States
| | - Brian Oliver
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaUnited States
| | - Julie A Brill
- Cell Biology Program, The Hospital for Sick ChildrenTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
- Institute of Medical Science, University of TorontoTorontoCanada
| | - Henry Krause
- Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Molecular Genetics, University of TorontoTorontoCanada
| | - Erika L Matunis
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | | | - Stephen DiNardo
- Department of Cell and Developmental Biology, The Perelman School of Medicine and The Penn Institute for Regenerative MedicinePhiladelphiaUnited States
| | - Margaret T Fuller
- Department of Developmental Biology, Stanford University School of MedicineStanfordUnited States
- Department of Genetics, Stanford UniversityStanfordUnited States
| |
Collapse
|
21
|
Hétié P, de Cuevas M, Matunis EL. The adult Drosophila testis lacks a mechanism to replenish missing niche cells. Development 2023; 150:dev201148. [PMID: 36503989 PMCID: PMC10110489 DOI: 10.1242/dev.201148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
The adult Drosophila testis contains a well-defined niche created by a cluster of hub cells, which secrete signals that maintain adjacent germline stem cells and somatic cyst stem cells (CySCs). Hub cells are normally quiescent in adult flies but can exit quiescence, delaminate from the hub and convert into CySCs after ablation of all CySCs. The opposite event, CySC conversion into hub cells, was proposed to occur under physiological conditions, but the frequency of this event is debated. Here, to probe further the question of whether or not hub cells can be regenerated, we developed methods to genetically ablate some or all hub cells. Surprisingly, when flies were allowed to recover from ablation, the missing hub cells were not replaced. Hub cells did not exit quiescence after partial ablation of hub cells, and labeled cells from outside the hub did not enter the hub during or after ablation. Despite its ability to exit quiescence in response to CySC ablation, we conclude that the hub in the adult Drosophila testis does not have a mechanism to replenish missing hub cells.
Collapse
Affiliation(s)
- Phylis Hétié
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Margaret de Cuevas
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Erika L. Matunis
- Department of Cell Biology, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
Abstract
Adult tissues in Metazoa dynamically remodel their structures in response to environmental challenges including sudden injury, pathogen infection, and nutritional fluctuation, while maintaining quiescence under homoeostatic conditions. This characteristic, hereafter referred to as adult tissue plasticity, can prevent tissue dysfunction and improve the fitness of organisms in continuous and/or severe change of environments. With its relatively simple tissue structures and genetic tools, studies using the fruit fly Drosophila melanogaster have provided insights into molecular mechanisms that control cellular responses, particularly during regeneration and nutrient adaptation. In this review, we present the current understanding of cellular mechanisms, stem cell proliferation, polyploidization, and cell fate plasticity, all of which enable adult tissue plasticity in various Drosophila adult organs including the midgut, the brain, and the gonad, and discuss the organismal strategy in response to environmental changes and future directions of the research.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Yu-Ichiro Nakajima
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Greenspan LJ, de Cuevas M, Le KH, Viveiros JM, Matunis EL. Activation of the EGFR/MAPK pathway drives transdifferentiation of quiescent niche cells to stem cells in the Drosophila testis niche. eLife 2022; 11:e70810. [PMID: 35468055 PMCID: PMC9038189 DOI: 10.7554/elife.70810] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 03/31/2022] [Indexed: 12/28/2022] Open
Abstract
Adult stem cells are maintained in niches, specialized microenvironments that regulate their self-renewal and differentiation. In the adult Drosophila testis stem cell niche, somatic hub cells produce signals that regulate adjacent germline stem cells (GSCs) and somatic cyst stem cells (CySCs). Hub cells are normally quiescent, but after complete genetic ablation of CySCs, they can proliferate and transdifferentiate into new CySCs. Here we find that Epidermal growth factor receptor (EGFR) signaling is upregulated in hub cells after CySC ablation and that the ability of testes to recover from ablation is inhibited by reduced EGFR signaling. In addition, activation of the EGFR pathway in hub cells is sufficient to induce their proliferation and transdifferentiation into CySCs. We propose that EGFR signaling, which is normally required in adult cyst cells, is actively inhibited in adult hub cells to maintain their fate but is repurposed to drive stem cell regeneration after CySC ablation.
Collapse
Affiliation(s)
- Leah J Greenspan
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Margaret de Cuevas
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Kathy H Le
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Graduate Program in Biophysics, Stanford UniversityStanfordUnited States
| | - Jennifer M Viveiros
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Erika L Matunis
- Department of Cell Biology, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
24
|
Anllo L, DiNardo S. Visceral mesoderm signaling regulates assembly position and function of the Drosophila testis niche. Dev Cell 2022; 57:1009-1023.e5. [PMID: 35390292 PMCID: PMC9050945 DOI: 10.1016/j.devcel.2022.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 02/07/2022] [Accepted: 03/14/2022] [Indexed: 12/20/2022]
Abstract
Tissue homeostasis often requires a properly placed niche to support stem cells. Morphogenetic processes that position a niche are just being described. For the Drosophila testis, we recently showed that pro-niche cells, specified at disparate positions during early gonadogenesis, must assemble into one collective at the anterior of the gonad. We now find that Slit and FGF signals emanating from adjacent visceral mesoderm regulate assembly. In response to signaling, niche cells express islet, which we find is also required for niche assembly. Without signaling, niche cells specified furthest from the anterior are unable to migrate, remaining dispersed. The function of such niches is severely disrupted, with niche cells evading cell cycle quiescence, compromised in their ability to signal the incipient stem cell pool, and failing to orient stem cell divisions properly. Our work identifies both extrinsic signaling and intrinsic responses required for proper assembly and placement of the testis niche.
Collapse
Affiliation(s)
- Lauren Anllo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States
| | - Stephen DiNardo
- Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| |
Collapse
|
25
|
To V, Kim HJ, Jang W, Sreejith P, Kim C. Lin28 and Imp are Required for Stability of Bowl Transcripts in Hub
Cells of the Drosophila Testis. Dev Reprod 2021; 25:313-319. [PMID: 35141457 PMCID: PMC8807131 DOI: 10.12717/dr.2021.25.4.313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/28/2021] [Accepted: 12/15/2021] [Indexed: 11/24/2022]
Abstract
Hub cells comprise a niche for germline stem cells and cyst stem cells in the
Drosophila testis. Hub cells arise from common somatic
gonadal precursors in embryos, but the mechanism of their specification is still
poorly understood. Here we find that RNA binding proteins Lin28 and Imp mediate
transcript stability of Bowl, a known hub specification factor; Bowl transcripts
were reduced in the testis of Lin28 and Imp mutants, and also when RNA-mediated
interference against Lin28 or Imp was expressed in hub cells. In tissue culture
Luciferase assays involving the Bowl 3’UTR, stability of Luc reporter
transcripts depended on the Bowl 3’UTR and required Lin28 and Imp. Our
findings suggest that proper Bowl function during hub cell specification
requires Lin28 and Imp in the testis hub cells.
Collapse
Affiliation(s)
- Van To
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | - Hyun Ju Kim
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | - Wijeong Jang
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
| | | | - Changsoo Kim
- School of Biological Sciences and
Technology, Chonnam National University, Gwangju
61186, Korea
- Corresponding author Changsoo Kim, School of
Biological Sciences and Technology, Chonnam National University, Gwangju 61186,
Korea. Tel: +82-62-530-5201, E-mail:
| |
Collapse
|
26
|
Herrera SC, Sainz de la Maza D, Grmai L, Margolis S, Plessel R, Burel M, O'Connor M, Amoyel M, Bach EA. Proliferative stem cells maintain quiescence of their niche by secreting the Activin inhibitor Follistatin. Dev Cell 2021; 56:2284-2294.e6. [PMID: 34363758 PMCID: PMC8387025 DOI: 10.1016/j.devcel.2021.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 05/14/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022]
Abstract
Aging causes stem cell dysfunction as a result of extrinsic and intrinsic changes. Decreased function of the stem cell niche is an important contributor to this dysfunction. We use the Drosophila testis to investigate what factors maintain niche cells. The testis niche comprises quiescent “hub” cells and supports two mitotic stem cell pools: germline stem cells and somatic cyst stem cells (CySCs). We identify the cell-cycle-responsive Dp/E2f1 transcription factor as a crucial non-autonomous regulator required in CySCs to maintain hub cell quiescence. Dp/E2f1 inhibits local Activin ligands through production of the Activin antagonist Follistatin (Fs). Inactivation of Dp/E2f1 or Fs in CySCs or promoting Activin receptor signaling in hub cells causes transdifferentiation of hub cells into fully functional CySCs. This Activin-dependent communication between CySCs and hub regulates the physiological decay of the niche with age and demonstrates that hub cell quiescence results from signals from surrounding stem cells. Dp/E2f1 is required in stem cells to non-autonomously maintain niche quiescence Dp/E2f1 promotes niche quiescence through Fs, an Activin antagonist Activin signaling in niche cells causes transdifferentiation into functional stem cells Fs in stem cells regulates the physiological decay of the niche with age
Collapse
Affiliation(s)
- Salvador C Herrera
- Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA; Centro Andaluz de Biología del Desarrollo, CSIC/Universidad Pablo de Olavide/JA, Carretera de Utrera km 1, 41013 Sevilla, Spain
| | - Diego Sainz de la Maza
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Lydia Grmai
- Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Shally Margolis
- Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Rebecca Plessel
- Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Michael Burel
- Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Michael O'Connor
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Marc Amoyel
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| | - Erika A Bach
- Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
27
|
Grmai L, Harsh S, Lu S, Korman A, Deb IB, Bach EA. Transcriptomic analysis of feminizing somatic stem cells in the Drosophila testis reveals putative downstream effectors of the transcription factor Chinmo. G3 (BETHESDA, MD.) 2021; 11:jkab067. [PMID: 33751104 PMCID: PMC8759813 DOI: 10.1093/g3journal/jkab067] [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: 01/18/2021] [Accepted: 02/24/2021] [Indexed: 11/12/2022]
Abstract
One of the best examples of sexual dimorphism is the development and function of the gonads, ovaries and testes, which produce sex-specific gametes, oocytes, and spermatids, respectively. The development of these specialized germ cells requires sex-matched somatic support cells. The sexual identity of somatic gonadal cells is specified during development and must be actively maintained during adulthood. We previously showed that the transcription factor Chinmo is required to ensure the male sexual identity of somatic support cells in the Drosophila melanogaster testis. Loss of chinmo from male somatic gonadal cells results in feminization: they transform from squamous to epithelial-like cells that resemble somatic cells in the female gonad but fail to properly ensheath the male germline, causing infertility. To identify potential target genes of Chinmo, we purified somatic cells deficient for chinmo from the adult Drosophila testis and performed next-generation sequencing to compare their transcriptome to that of control somatic cells. Bioinformatics revealed 304 and 1549 differentially upregulated and downregulated genes, respectively, upon loss of chinmo in early somatic cells. Using a combination of methods, we validated several differentially expressed genes. These data sets will be useful resources to the community.
Collapse
Affiliation(s)
- Lydia Grmai
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Sneh Harsh
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Sean Lu
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Aryeh Korman
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Ishan B Deb
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| | - Erika A Bach
- Department of Biochemistry & Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY
| |
Collapse
|
28
|
Li YR, Lai HW, Huang HH, Chen HC, Fugmann SD, Yang SY. Trajectory mapping of the early Drosophila germline reveals controls of zygotic activation and sex differentiation. Genome Res 2021; 31:1011-1023. [PMID: 33858841 PMCID: PMC8168578 DOI: 10.1101/gr.271148.120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 04/07/2021] [Indexed: 01/29/2023]
Abstract
Germ cells in Drosophila melanogaster are specified maternally shortly after fertilization and are transcriptionally quiescent until their zygotic genome is activated to sustain further development. To understand the molecular basis of this process, we analyzed the progressing transcriptomes of early male and female germ cells at the single-cell level between germline specification and coalescence with somatic gonadal cells. Our data comprehensively cover zygotic activation in the germline genome, and analyses on genes that exhibit germline-restricted expression reveal that polymerase pausing and differential RNA stability are important mechanisms that establish gene expression differences between the germline and soma. In addition, we observe an immediate bifurcation between the male and female germ cells as zygotic transcription begins. The main difference between the two sexes is an elevation in X Chromosome expression in females relative to males, signifying incomplete dosage compensation, with a few select genes exhibiting even higher expression increases. These indicate that the male program is the default mode in the germline that is driven to female development with a second X Chromosome.
Collapse
Affiliation(s)
- Yi-Ru Li
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan
| | - Hsiao Wen Lai
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan
| | - Hsiao Han Huang
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan
| | - Hsing-Chun Chen
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan
| | - Sebastian D Fugmann
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan.,Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan.,Department of Nephrology, Linkou Chang Gung Memorial Hospital, Kweishan, Taoyuan 333 Taiwan
| | - Shu Yuan Yang
- Department and College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan.,Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Kweishan, Taoyuan 333 Taiwan.,Department of Gynecology, Linkou Chang Gung Memorial Hospital, Kweishan, Taoyuan 333 Taiwan
| |
Collapse
|
29
|
Zhou H, Whitworth C, Pozmanter C, Neville MC, Van Doren M. Doublesex regulates fruitless expression to promote sexual dimorphism of the gonad stem cell niche. PLoS Genet 2021; 17:e1009468. [PMID: 33788836 PMCID: PMC8041189 DOI: 10.1371/journal.pgen.1009468] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 04/12/2021] [Accepted: 03/04/2021] [Indexed: 11/25/2022] Open
Abstract
Doublesex (Dsx) and Fruitless (Fru) are the two downstream transcription factors that actuate Drosophila sex determination. While Dsx assists Fru to regulate sex-specific behavior, whether Fru collaborates with Dsx in regulating other aspects of sexual dimorphism remains unknown. One important aspect of sexual dimorphism is found in the gonad stem cell (GSC) niches, where male and female GSCs are regulated to create large numbers of sperm and eggs. Here we report that Fru is expressed male-specifically in the GSC niche and plays important roles in the development and maintenance of these cells. Unlike previously-studied aspects of sex-specific Fru expression, which are regulated by Transformer (Tra)-mediated alternative splicing, we show that male-specific expression of fru in the gonad is regulated downstream of dsx, and is independent of tra. fru genetically interacts with dsx to support maintenance of the niche throughout development. Ectopic expression of fru inhibited female niche formation and partially masculinized the ovary. fru is also required autonomously for cyst stem cell maintenance and cyst cell survival. Finally, we identified a conserved Dsx binding site upstream of fru promoter P4 that regulates fru expression in the niche, indicating that fru is likely a direct target for transcriptional regulation by Dsx. These findings demonstrate that fru acts outside the nervous system to influence sexual dimorphism and reveal a new mechanism for regulating sex-specific expression of fru that is regulated at the transcriptional level by Dsx, rather than by alternative splicing by Tra.
Collapse
Affiliation(s)
- Hong Zhou
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, United States of America
| | - Cale Whitworth
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, United States of America
| | - Caitlin Pozmanter
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, United States of America
| | - Megan C. Neville
- Centre for Neural Circuits and Behaviour, University of Oxford, Tinsley Building, Mansfield Road, Oxford, United Kingdom
| | - Mark Van Doren
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, United States of America
| |
Collapse
|
30
|
Nandi A, Chowdhuri DK. Cadmium mediated redox modulation in germline stem cells homeostasis affects reproductive health of Drosophila males. JOURNAL OF HAZARDOUS MATERIALS 2021; 402:123737. [PMID: 33254766 DOI: 10.1016/j.jhazmat.2020.123737] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 06/12/2023]
Abstract
Maintenance of male germline stem cells (GSCs) homeostasis is crucial for successful reproductive life of adults. New insights gained on dysfunction in stem cell maintenance could be the basis of stem cell dependent ailment during adulthood. Cadmium (Cd), a reported male reproductive toxicant, has been explored inadequately for its impact on male GSCs maintenance. The present study, therefore, has been aimed to evaluate the adverse effect of Cd on the homeostasis of GSCs by using Drosophila testis as an in vivo model. Following developmental exposure of environmentally relevant concentrations of Cd (5.0, 10.0 and 20.0 μg/mL) to Drosophila, we showed that a significantly increased level of reactive oxygen species (ROS) at 20.0 μg/mL of Cd resulted in alteration of GSCs number accompanied by inappropriate differentiation leading to reduced sperm number and eventually poor reproductive performance in exposed organism. Rescuing effect was evident by overexpressing sod in the early germ cell stage. The study suggests that an alteration in GSCs homeostasis due to redox imbalance plays a pivotal role in Cd induced failure in male fertility. The study further advocates for the use of Drosophila as an alternative animal model for in vivo evaluation of male GSCs toxicity with minimal ethical concern.
Collapse
Affiliation(s)
- Abik Nandi
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, Uttar Pradesh, India
| | - Debapratim Kar Chowdhuri
- Embryotoxicology Laboratory, Environmental Toxicology Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, Uttar Pradesh, India.
| |
Collapse
|
31
|
Nelson KA, Warder BN, DiNardo S, Anllo L. Dissection and Live-Imaging of the Late Embryonic Drosophila Gonad. J Vis Exp 2020:10.3791/61872. [PMID: 33135688 PMCID: PMC7878017 DOI: 10.3791/61872] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
The Drosophila melanogaster male embryonic gonad is an advantageous model to study various aspects of developmental biology including, but not limited to, germ cell development, piRNA biology, and niche formation. Here, we present a dissection technique to live-image the gonad ex vivo during a period when in vivo live-imaging is highly ineffective. This protocol outlines how to transfer embryos to an imaging dish, choose appropriately-staged male embryos, and dissect the gonad from its surrounding tissue while still maintaining its structural integrity. Following dissection, gonads can be imaged using a confocal microscope to visualize dynamic cellular processes. The dissection procedure requires precise timing and dexterity, but we provide insight on how to prevent common mistakes and how to overcome these challenges. To our knowledge this is the first dissection protocol for the Drosophila embryonic gonad, and will permit live-imaging during an otherwise inaccessible window of time. This technique can be combined with pharmacological or cell-type specific transgenic manipulations to study any dynamic processes occurring within or between the cells in their natural gonadal environment.
Collapse
Affiliation(s)
- Kara A. Nelson
- Cell and Developmental Biology Department and the Penn Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Bailey N. Warder
- Cell and Developmental Biology Department and the Penn Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Stephen DiNardo
- Cell and Developmental Biology Department and the Penn Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania
| | - Lauren Anllo
- Cell and Developmental Biology Department and the Penn Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania;
| |
Collapse
|
32
|
Greenspan LJ, Matunis EL. Retinoblastoma Intrinsically Regulates Niche Cell Quiescence, Identity, and Niche Number in the Adult Drosophila Testis. Cell Rep 2019; 24:3466-3476.e8. [PMID: 30257208 PMCID: PMC6226258 DOI: 10.1016/j.celrep.2018.08.083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/29/2018] [Accepted: 08/27/2018] [Indexed: 12/20/2022] Open
Abstract
Homeostasis in adult tissues depends on the precise regulation of stem cells and their surrounding microenvironments, or niches. Here, we show that the cell cycle inhibitor and tumor suppressor Retinoblastoma (RB) is a critical regulator of niche cells in the Drosophila testis. The testis contains a single niche, composed of somatic hub cells, that signals to adjacent germline and somatic stem cells. Hub cells are normally quiescent, but knockdown of the RB homolog Rbf in these cells causes them to proliferate and convert to somatic stem cells. Over time, mutant hub cell clusters enlarge and split apart, forming ectopic hubs surrounded by active stem cells. Furthermore, we show that Rbf’s ability to restrict niche number depends on the transcription factors E2F and Escargot and the adhesion molecule E-cadherin. Together this work reveals how precise modulation of niche cells, not only the stem cells they support, can drive regeneration and disease. Greenspan and Matunis find that the tumor suppressor Retinoblastoma is required in niche cells to maintain quiescence, cell fate, and niche number. Loss of Retinoblastoma causes niche cell divisions, conversion to somatic stem cells, and ectopic niche formation through niche fission, suggesting that mutations in niche cells may drive disease.
Collapse
Affiliation(s)
- Leah J Greenspan
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Erika L Matunis
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
33
|
Participation of the Serine Protease Jonah66Ci in the Drosophila Antinematode Immune Response. Infect Immun 2019; 87:IAI.00094-19. [PMID: 31182620 DOI: 10.1128/iai.00094-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/29/2019] [Indexed: 01/27/2023] Open
Abstract
Serine proteases and serine protease homologs form the second largest gene family in the Drosophila melanogaster genome. Certain genes in the Jonah multigene family encoding serine proteases have been implicated in the fly antiviral immune response. Here, we report the involvement of Jonah66Ci in the Drosophila immune defense against Steinernema carpocapsae nematode infection. We find that Drosophila Jonah66Ci is upregulated in response to symbiotic (carrying the mutualistic bacterium Xenorhabdus nematophila) or axenic (lacking Xenorhabdus) Steinernema nematodes and is expressed exclusively in the gut of Drosophila larvae. Inactivation of Jonah66Ci provides a survival advantage to larvae against axenic nematodes and results in differential expression of Toll and Imd pathway effector genes, specifically in the gut. Also, inactivation of Jonah66Ci increases the numbers of enteroendocrine and mitotic cells in the gut of uninfected larvae, and infection with Steinernema nematodes reduces their numbers, whereas the numbers of intestinal stem cells are unaffected by nematode infection. Jonah66Ci knockdown further reduces nitric oxide levels in response to infection with symbiotic Steinernema nematodes. Finally, we show that Jonah66Ci knockdown does not alter the feeding rates of uninfected Drosophila larvae; however, infection with axenic Steinernema nematodes lowers larval feeding. In conclusion, we report that Jonah66Ci participates in maintaining homeostasis of certain physiological processes in Drosophila larvae in the context of Steinernema nematode infection. Similar findings will take us a step further toward understanding the molecular and physiological mechanisms that take place during parasitic nematode infection in insects.
Collapse
|
34
|
Camara N, Whitworth C, Dove A, Van Doren M. Doublesex controls specification and maintenance of the gonad stem cell niches in Drosophila. Development 2019; 146:dev.170001. [PMID: 31043421 DOI: 10.1242/dev.170001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 04/23/2019] [Indexed: 11/20/2022]
Abstract
Sex-specific development of the gonads is a key aspect of sexual dimorphism that is regulated by Doublesex/Mab3-related transcription factors (DMRTs) in diverse animal species. We find that in mutants for Drosophila dsx, important components of the male and female gonad stem cell niches (hubs and terminal filaments/cap cells, respectively) still form. Initially, gonads in all dsx mutants (both XX and XY) initiate the male program of development, but later half of these gonads switch to form female stem cell niche structures. One individual can have both male-type and female-type gonad niches; however, male and female niches are usually not observed in the same gonad, indicating that cells make a 'group decision' about which program to follow. We conclude that dsx does not act in an instructive manner to regulate male versus female niche formation, as these structures form in the absence of dsx function. Instead, dsx acts to 'tip the balance' between the male or female programs, which are then executed independently of dsx We show that bric a brac acts downstream of dsx to control the male versus female niche decision. These results indicate that, in both flies and mammals, the sexual fate of the somatic gonad is remarkably plastic and is controlled by a combination of autonomous and non-autonomous cues.
Collapse
Affiliation(s)
- Nicole Camara
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Cale Whitworth
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Abigail Dove
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| | - Mark Van Doren
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD 21218, USA
| |
Collapse
|
35
|
Lu Y, Yao Y, Li Z. Ectopic Dpp signaling promotes stem cell competition through EGFR signaling in the Drosophila testis. Sci Rep 2019; 9:6118. [PMID: 30992503 PMCID: PMC6467874 DOI: 10.1038/s41598-019-42630-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 04/02/2019] [Indexed: 12/28/2022] Open
Abstract
Stem cell competition could select the fittest stem cells and potentially control tumorigenesis. However, little is known about the underlying molecular mechanisms. Here, we find that ectopic Decapentaplegic (Dpp) signal activation by expressing a constitutively active form of Thickveins (TkvCA) in cyst stem cells (CySCs) leads to competition between CySCs and germline stem cells (GSCs) for niche occupancy and GSC loss. GSCs are displaced from the niche and undergo differentiation. Interestingly, we find that induction of TkvCA results in elevated expression of vein, which further activates Epidermal Growth Factor Receptor (EGFR) signaling in CySCs to promote their proliferation and compete GSCs out of the niche. Our findings elucidate the important role of Dpp signaling in regulating stem cell competition and tumorigenesis, which could be shed light on tumorigenesis and cancer treatment in mammals.
Collapse
Affiliation(s)
- Yanfen Lu
- College of Plant Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Changping District, Beijing, 102206, China
| | - Yuncong Yao
- College of Plant Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Changping District, Beijing, 102206, China.
| | - Zhouhua Li
- College of Life Sciences, Capital Normal University, Beijing, 100048, China.
| |
Collapse
|
36
|
Anllo L, Plasschaert LW, Sui J, DiNardo S. Live imaging reveals hub cell assembly and compaction dynamics during morphogenesis of the Drosophila testis niche. Dev Biol 2018; 446:102-118. [PMID: 30553808 DOI: 10.1016/j.ydbio.2018.12.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/19/2018] [Accepted: 12/12/2018] [Indexed: 01/23/2023]
Abstract
Adult stem cells are often found in specialized niches, where the constituent cells direct self-renewal of their stem cell pool. The niche is therefore crucial for both normal homeostasis and tissue regeneration. In many mammalian tissues, niche cells have classically been difficult to identify, which has hampered any understanding of how tissues first construct niches during development. Fortunately, the Drosophila germline stem cell (GSC) niche is well defined, allowing for unambiguous identification of both niche cells and resident stem cells. The testis niche first forms in the early embryo, during a late stage of gonadogenesis. Here, using live-imaging both in vivo and ex vivo, we follow pro-niche cells as they assemble and assume their final form. We show that after ex vivo culture the niche appears fully functional, as judged by enrichment of adhesion proteins, the ability to activate STAT in adjacent GSCs, and to direct GSCs to divide orthogonally to the niche, just as they would in situ. Collectively, our imaging has generated several novel insights on niche morphogenesis that could not be inferred from fixed images alone. We identify dynamic processes that constitute an assembly phase and a compaction phase during morphogenesis. The compaction phase correlates with cell neighbor exchange among the assembled pro-niche cells, as well as a burst of divisions among newly recruited stem cells. Before compaction, an assembly phase involves the movement of pro-niche cells along the outer periphery of the gonad, using the extracellular matrix (ECM) to assemble at the anterior of the gonad. Finally, live-imaging in integrin mutants allows us to define the role of pro-niche cell-ECM interaction with regard to the new assembly and compaction dynamics revealed here.
Collapse
Affiliation(s)
- Lauren Anllo
- The Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| | - Lindsey W Plasschaert
- The Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| | - Justin Sui
- The Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| | - Stephen DiNardo
- The Perelman School of Medicine at the University of Pennsylvania, 421 Curie Blvd, Philadelphia, PA 19104, United States; The Penn Institute for Regenerative Medicine, 421 Curie Blvd, Philadelphia, PA 19104, United States.
| |
Collapse
|
37
|
Albert EA, Puretskaia OA, Terekhanova NV, Labudina A, Bökel C. Direct control of somatic stem cell proliferation factors by the Drosophila testis stem cell niche. Development 2018; 145:dev.156315. [PMID: 30002131 DOI: 10.1242/dev.156315] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 07/06/2018] [Indexed: 12/16/2022]
Abstract
Niches have traditionally been characterised as signalling microenvironments that allow stem cells to maintain their fate. This definition implicitly assumes that the various niche signals are integrated towards a binary fate decision between stemness and differentiation. However, observations in multiple systems have demonstrated that stem cell properties, such as proliferation and self-renewal, can be uncoupled at the level of niche signalling input, which is incompatible with this simplified view. We have studied the role of the transcriptional regulator Zfh1, a shared target of the Hedgehog and Jak/Stat niche signalling pathways, in the somatic stem cells of the Drosophila testis. We found that Zfh1 binds and downregulates salvador and kibra, two tumour suppressor genes of the Hippo/Wts/Yki pathway, thereby restricting Yki activation and proliferation to the Zfh1+ stem cells. These observations provide an unbroken link from niche signal input to an individual aspect of stem cell behaviour that does not, at any step, involve a fate decision. We discuss the relevance of these findings for an overall concept of stemness and niche function.
Collapse
Affiliation(s)
- Eugene A Albert
- Centre for Regenerative Therapies Dresden, Technical University Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Olga A Puretskaia
- Centre for Regenerative Therapies Dresden, Technical University Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Nadezhda V Terekhanova
- Sector for Molecular Evolution, Institute for Information Transmission Problems of the RAS (Kharkevich Institute), Moscow 127994, Russia.,N. K. Koltsov Institute of Developmental Biology of the RAS, Moscow 119334, Russia.,Laboratory of Molecular Genetics, Russian Federal Research Institute of Fisheries and Oceanography, Moscow 107140, Russia
| | - Anastasia Labudina
- Centre for Regenerative Therapies Dresden, Technical University Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Christian Bökel
- Centre for Regenerative Therapies Dresden, Technical University Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| |
Collapse
|
38
|
Spermiogenesis and Male Fertility Require the Function of Suppressor of Hairy-Wing in Somatic Cyst Cells of Drosophila. Genetics 2018; 209:757-772. [PMID: 29739818 DOI: 10.1534/genetics.118.301088] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/01/2018] [Indexed: 02/07/2023] Open
Abstract
Drosophila Suppressor of Hairy-wing [Su(Hw)] protein is an example of a multivalent transcription factor. Although best known for its role in establishing the chromatin insulator of the gypsy retrotransposon, Su(Hw) functions as an activator and repressor at non-gypsy genomic sites. It remains unclear how the different regulatory activities of Su(Hw) are utilized during development. Motivated from observations of spatially restricted expression of Su(Hw) in the testis, we investigated the role of Su(Hw) in spermatogenesis to advance an understanding of its developmental contributions as an insulator, repressor, and activator protein. We discovered that Su(Hw) is required for sustained male fertility. Although dynamics of Su(Hw) expression coincide with changes in nuclear architecture and activation of coregulated testis-specific gene clusters, we show that loss of Su(Hw) does not disrupt meiotic chromosome pairing or transcription of testis-specific genes, suggesting that Su(Hw) has minor architectural or insulator functions in the testis. Instead, Su(Hw) has a prominent role as a repressor of neuronal genes, consistent with suggestions that Su(Hw) is a functional homolog of mammalian REST, a repressor of neuronal genes in non-neuronal tissues. We show that Su(Hw) regulates transcription in both germline and somatic cells. Surprisingly, the essential spermatogenesis function of Su(Hw) resides in somatic cyst cells, implying context-specific consequences due to loss of this transcription factor. Together, our studies highlight that Su(Hw) has a major developmental function as a transcriptional repressor, with the effect of its loss dependent upon the cell-specific factors.
Collapse
|
39
|
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
|
40
|
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
|
41
|
Regulatory Mechanisms of the Germline Stem Cell Niche in Drosophila melanogaster. DIVERSITY AND COMMONALITY IN ANIMALS 2018. [DOI: 10.1007/978-4-431-56609-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
42
|
miR-9a modulates maintenance and ageing of Drosophila germline stem cells by limiting N-cadherin expression. Nat Commun 2017; 8:600. [PMID: 28928361 PMCID: PMC5605507 DOI: 10.1038/s41467-017-00485-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 07/03/2017] [Indexed: 12/19/2022] Open
Abstract
Ageing is characterized by a decline in stem cell functionality leading to dampened tissue regeneration. While the expression of microRNAs across multiple species is markedly altered with age, the mechanism by which they govern stem cell-sustained tissue regeneration is unknown. We report that in the Drosophila testis, the conserved miR-9a is expressed in germline stem cells and its levels are significantly elevated during ageing. Transcriptome and functional analyses show that miR-9a directly regulates the expression of the adhesion molecule N-cadherin (N-cad). miR-9a null mutants maintain a higher number of stem cells even in the aged tissue. Remarkably, this rise fails to improve tissue regeneration and results in reduced male fertility. Similarly, overexpression of N-cad also results in elevated stem cell number and decreased regeneration. We propose that miR-9a downregulates N-cad to enable adequate detachment of stem cells toward differentiation, thus providing the necessary directionality toward terminal differentiation and spermatogenesis.In the Drosophila testis, ageing leads to loss of germline stem cells. Here, the authors show that, during ageing in Drosophila, miR-9a is upregulated in male germline stem cells and regulates their proliferation by targeting N-cadherin.
Collapse
|
43
|
A genetically enhanced sterile insect technique against the fruit fly, Bactrocera dorsalis (Hendel) by feeding adult double-stranded RNAs. Sci Rep 2017. [PMID: 28642479 PMCID: PMC5481416 DOI: 10.1038/s41598-017-04431-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
RNAi based sterile insect technique (SIT) is an authentic insect management approach but requires proper target genes. During this study, spermless males were developed by interfering with germ cell differentiation and azoospermia related genes. Data demonstrates significant reductions in the target genes expressions (boul, zpg, dsxM, fzo and gas8) after oral dsRNAs administration. Knock down of target genes significantly affected the reproductive ability of males and reduced egg-hatching as compared to the control group. Furthermore, different combinations of selected gene dsRNAs (boul + zpg, boul + dsxM and zpg + dsxM) were made, which resulted up to 85.40% of male sterility. The most effective combination was selected to prepare different concentrations of dsRNA, 250, 500, 750 and 1000 ng/μl, that caused 18.97%, 38.68%, 58.02% and 85.40% male sterility, respectively. Subsequently, 1000 ng/μl of the same combination of ds-RNAs was used against differently aged adult flies (1, 5, 7, 10 days) which lead to 85.40%, 31.42%, 21.76% and 9.90% male sterility, respectively. SIT developed in this study showed that, boul + zpg combination of dsRNA feeding for 6 hours significantly reduced the number of spermatozoa and viability of sperm in 1-day-old B. dorsalis flies. In short, this study provides an effective SIT technique for long-term B. dorsalis management.
Collapse
|
44
|
Siddall NA, Hime GR. A Drosophila toolkit for defining gene function in spermatogenesis. Reproduction 2017; 153:R121-R132. [PMID: 28073824 DOI: 10.1530/rep-16-0347] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 12/15/2016] [Accepted: 01/10/2017] [Indexed: 12/29/2022]
Abstract
Expression profiling and genomic sequencing methods enable the accumulation of vast quantities of data that relate to the expression of genes during the maturation of male germ cells from primordial germ cells to spermatozoa and potential mutations that underlie male infertility. However, the determination of gene function in specific aspects of spermatogenesis or linking abnormal gene function with infertility remain rate limiting, as even in an era of CRISPR analysis of gene function in mammalian models, this still requires considerable resources and time. Comparative developmental biology studies have shown the remarkable conservation of spermatogenic developmental processes from insects to vertebrates and provide an avenue of rapid assessment of gene function to inform the potential roles of specific genes in rodent and human spermatogenesis. The vinegar fly, Drosophila melanogaster, has been used as a model organism for developmental genetic studies for over one hundred years, and research with this organism produced seminal findings such as the association of genes with chromosomes, the chromosomal basis for sexual identity, the mutagenic properties of X-irradiation and the isolation of the first tumour suppressor mutations. Drosophila researchers have developed an impressive array of sophisticated genetic techniques for analysis of gene function and genetic interactions. This review focuses on how these techniques can be utilised to study spermatogenesis in an organism with a generation time of 9 days and the capacity to introduce multiple mutant alleles into an individual organism in a relatively short time frame.
Collapse
Affiliation(s)
- N A Siddall
- Department of Anatomy and NeuroscienceThe University of Melbourne, Parkville, Victoria, Australia
| | - G R Hime
- Department of Anatomy and NeuroscienceThe University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
45
|
Fairchild MJ, Yang L, Goodwin K, Tanentzapf G. Occluding Junctions Maintain Stem Cell Niche Homeostasis in the Fly Testes. Curr Biol 2016; 26:2492-2499. [PMID: 27546574 DOI: 10.1016/j.cub.2016.07.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/23/2016] [Accepted: 07/08/2016] [Indexed: 12/16/2022]
Abstract
Stem cells can be controlled by their local microenvironment, known as the stem cell niche. The Drosophila testes contain a morphologically distinct niche called the hub, composed of a cluster of between 8 and 20 cells known as hub cells, which contact and regulate germline stem cells (GSCs) and somatic cyst stem cells (CySCs). Both hub cells and CySCs originate from somatic gonadal precursor cells during embryogenesis, but whereas hub cells, once specified, cease all mitotic activity, CySCs remain mitotic into adulthood [1, 2]. Cyst cells, derived from the CySCs, first encapsulate the germline and then, using occluding junctions, form an isolating permeability barrier [3]. This barrier promotes germline differentiation by excluding niche-derived stem cell maintenance factors. Here, we show that the somatic permeability barrier is also required to regulate stem cell niche homeostasis. Loss of occluding junction components in the somatic cells results in hub overgrowth. Enlarged hubs are active and recruit more GSCs and CySCs to the niche. Surprisingly, hub growth results from depletion of occluding junction components in cyst cells, not from depletion in the hub cells themselves. Moreover, hub growth is caused by incorporation of cells that previously expressed markers for cyst cells and not by hub cell proliferation. Importantly, depletion of occluding junctions disrupts Notch and mitogen-activated protein kinase (MAPK) signaling, and hub overgrowth defects are partially rescued by modulation of either signaling pathway. Overall, these data show that occluding junctions shape the signaling environment between the soma and the germline in order to maintain niche homeostasis.
Collapse
Affiliation(s)
- Michael J Fairchild
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Lulu Yang
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Katharine Goodwin
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
46
|
Lee JY, Chen JY, Shaw JL, Chang KT. Maintenance of Stem Cell Niche Integrity by a Novel Activator of Integrin Signaling. PLoS Genet 2016; 12:e1006043. [PMID: 27191715 PMCID: PMC4871447 DOI: 10.1371/journal.pgen.1006043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 04/19/2016] [Indexed: 01/22/2023] Open
Abstract
Stem cells depend critically on the surrounding microenvironment, or niche, for their maintenance and self-renewal. While much is known about how the niche regulates stem cell self-renewal and differentiation, mechanisms for how the niche is maintained over time are not well understood. At the apical tip of the Drosophila testes, germline stem cells (GSCs) and somatic stem cells share a common niche formed by hub cells. Here we demonstrate that a novel protein named Shriveled (Shv) is necessary for the maintenance of hub/niche integrity. Depletion of Shv protein results in age-dependent deterioration of the hub structure and loss of GSCs, whereas upregulation of Shv preserves the niche during aging. We find Shv is a secreted protein that modulates DE-cadherin levels through extracellular activation of integrin signaling. Our work identifies Shv as a novel activator of integrin signaling and suggests a new integration model in which crosstalk between integrin and DE-cadherin in niche cells promote their own preservation by maintaining the niche architecture. Stem cells are vital for development and for regeneration and repair of tissues in an organism. The ability of adult stem cells to maintain their “stemness” depends critically on the localized microenvironment, or niche. While much is known about how the niche regulates stem cell self-renewal and differentiation, mechanisms for how the niche is maintained during aging are not well understood. Using Drosophila testis as a model system, here we demonstrate that a protein we named Shriveled is a secreted protein that activates integrin signaling to preserve niche architecture. We also show that Shriveled-dependent activation of integrin maintains normal E-cadherin levels in the niche cells, providing a mechanism for niche maintenance. Interestingly, upregulation of Shriveled retards the loss of niche and stem cells seen during normal aging. Together, our work identifies Shriveled as a novel molecule required for preservation of the niche structure in the Drosophila testis.
Collapse
Affiliation(s)
- Joo Yeun Lee
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, United States of America
| | - Jessica Y. Chen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Jillian L. Shaw
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, United States of America
| | - Karen T. Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- Neuroscience Graduate Program, University of Southern California, Los Angeles, California, United States of America
- Department of Cell and Neurobiology, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Barton LJ, Lovander KE, Pinto BS, Geyer PK. Drosophila male and female germline stem cell niches require the nuclear lamina protein Otefin. Dev Biol 2016; 415:75-86. [PMID: 27174470 DOI: 10.1016/j.ydbio.2016.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 04/28/2016] [Accepted: 05/02/2016] [Indexed: 12/11/2022]
Abstract
The nuclear lamina is an extensive protein network that underlies the inner nuclear envelope. This network includes the LAP2-emerin-MAN1-domain (LEM-D) protein family, proteins that share an association with the chromatin binding protein Barrier-to-autointegration factor (BAF). Loss of individual LEM-D proteins causes progressive, tissue-restricted diseases, known as laminopathies. Mechanisms associated with laminopathies are not yet understood. Here we present our studies of one of the Drosophila nuclear lamina LEM-D proteins, Otefin (Ote), a homologue of emerin. Previous studies have shown that Ote is autonomously required for the survival of female germline stem cells (GSCs). We demonstrate that Ote is also required for survival of somatic cells in the ovarian niche, with loss of Ote causing a decrease in cap cell number and altered signal transduction. We show germ cell-restricted expression of Ote rescues these defects, revealing a non-autonomous function for Ote in niche maintenance and emphasizing that GSCs contribute to the maintenance of their own niches. Further, we investigate the requirement of Ote in the male fertility. We show that ote mutant males become prematurely sterile as they age. Parallel to observations in females, this sterility is associated with GSC loss and changes in somatic cells of the niche, phenotypes that are largely rescued by germ cell-restricted Ote expression. Taken together, our studies demonstrate that Ote is required autonomously for survival of two stem cell populations, as well as non-autonomously for maintenance of two somatic niches. Finally, our data add to growing evidence that LEM-D proteins have critical roles in stem cell survival and tissue homeostasis.
Collapse
Affiliation(s)
- Lacy J Barton
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA
| | - Kaylee E Lovander
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA
| | - Belinda S Pinto
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA
| | - Pamela K Geyer
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
48
|
Loza-Coll MA, Jones DL. Simultaneous control of stemness and differentiation by the transcription factor Escargot in adult stem cells: How can we tease them apart? Fly (Austin) 2016; 10:53-9. [PMID: 27077690 DOI: 10.1080/19336934.2016.1176650] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The homeostatic turnover of adult organs and their regenerative capacity following injury depend on a careful balance between stem cell self-renewal (to maintain or enlarge the stem cell pool) and differentiation (to replace lost tissue). We have recently characterized the role of the Drosophila Snail family transcription factor escargot (esg) in testis cyst stem cells (CySCs) (1,2) and intestinal stem cells (ISCs). (3,4) CySCs mutant for esg are not maintained as stem cells, but they remain capable of differentiating normally along the cyst cell lineage. In contrast, esg mutant CySCs that give rise to a closely related lineage, the apical hub cells, cannot maintain hub cell identity. Similarly, Esg maintains stemness of ISCs while regulating the terminal differentiation of progenitor cells into absorptive enterocytes or secretory enteroendocrine cells. Therefore, our findings suggest that Esg may play a conserved and pivotal regulatory role in adult stem cells, controlling both their maintenance and terminal differentiation. Here we propose that this dual regulatory role is due to simultaneous control by Esg of overlapping genetic programs and discuss the exciting challenges and opportunities that lie ahead to explore the underlying mechanisms experimentally.
Collapse
Affiliation(s)
- Mariano A Loza-Coll
- a Department of Biology , California State University , Northridge , CA , USA
| | - D Leanne Jones
- b Molecular, Cell and Developmental Biology, University of California , Los Angeles , CA , USA.,c Eli and Edythe Broad Center of Regenerative Medicine, University of California , Los Angeles , CA , USA
| |
Collapse
|
49
|
Monahan AJ, Starz-Gaiano M. Apontic regulates somatic stem cell numbers in Drosophila testes. BMC DEVELOPMENTAL BIOLOGY 2016; 16:5. [PMID: 26993259 PMCID: PMC4799534 DOI: 10.1186/s12861-016-0103-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 02/10/2016] [Indexed: 12/27/2022]
Abstract
BACKGROUND Microenvironments called niches maintain resident stem cell populations by balancing self-renewal with differentiation, but the genetic regulation of this process is unclear. The niche of the Drosophila testis is well-characterized and genetically tractable, making it ideal for investigating the molecular regulation of stem cell biology. The JAK/STAT pathway, activated by signals from a niche component called the hub, maintains both germline and somatic stem cells. RESULTS This study investigated the molecular regulation of the JAK/STAT pathway in the stem cells of the Drosophila testis. We determined that the transcriptional regulator Apontic (Apt) acts in the somatic (cyst) stem cells (CySCs) to balance differentiation and maintenance. We found Apt functions as a negative feedback inhibitor of STAT activity, which enables cyst cell maturation. Simultaneous loss of the STAT regulators apt and Socs36E, or the Stat92E-targeting microRNA miR-279, expanded the somatic stem cell-like population. CONCLUSIONS Genetic analysis revealed that a conserved genetic regulatory network limits JAK/STAT activity in the somatic stem cells of Drosophila testis. In these cells, we determined JAK/STAT signaling promotes apt expression. Then, Apt functions through Socs36E and miR-279 to attenuate pathway activation, which is required for timely CySC differentiation. We propose that Apt acts as a core component of a STAT-regulatory circuit to prevent stem cell overpopulation and allow stem cell maturation.
Collapse
Affiliation(s)
- Amanda J Monahan
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA.,Present Address: Department of Medicine, Division of Infectious Disease, University of Massachusetts Medical School, Worcester, MA, 01655, USA
| | - Michelle Starz-Gaiano
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA.
| |
Collapse
|
50
|
Abstract
The embryonic gonad of Drosophila melanogaster begins to display sexually dimorphic traits soon after its formation. Here we demonstrate the involvement of a wnt family ligand, wnt-2, in the induction of these sex-specific differences. We show that wnt-2 contributes to the survival of a male-specific population of somatic gonadal precursor cells (SGPs), the male-specific SGPs that are located at the posterior of the male gonad. We also show that the Wnt-2 ligand synergizes with the JAK-STAT ligand Upd, which is produced by SGPs at the anterior of the gonad to activate the STAT pathway in male germ cells. We suggest that the use of two spatially separated signaling systems to initiate the JAK-STAT stem cell maintenance pathway in germ cells provides a mechanism for increasing the pool of potential progenitors of the germline stem cells in the adult testes. Finally, we present evidence indicating that, like the JAK-STAT pathway, wnt-2 stimulates germ cells in male embryos to re-enter the cell cycle.
Collapse
|