Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters











Publication year range
1.
Development ; 150(20)2023 10 15.
Article in English | MEDLINE | ID: mdl-37795747

ABSTRACT

Obesity is linked to reduced fertility in various species, from Drosophila to humans. Considering that obesity is often induced by changes in diet or eating behavior, it remains unclear whether obesity, diet, or both reduce fertility. Here, we show that Drosophila females on a high-sugar diet become rapidly obese and less fertile as a result of increased death of early germline cysts and vitellogenic egg chambers (or follicles). They also have high glycogen, glucose and trehalose levels and develop insulin resistance in their fat bodies (but not ovaries). By contrast, females with adipocyte-specific knockdown of the anti-obesity genes brummer or adipose are obese but have normal fertility. Remarkably, females on a high-sugar diet supplemented with a separate source of water have mostly normal fertility and glucose levels, despite persistent obesity, high glycogen and trehalose levels, and fat body insulin resistance. These findings demonstrate that a high-sugar diet affects specific processes in oogenesis independently of insulin resistance, that high glucose levels correlate with reduced fertility on a high-sugar diet, and that obesity alone does not impair fertility.


Subject(s)
Drosophila melanogaster , Insulin Resistance , Animals , Humans , Female , Drosophila melanogaster/genetics , Trehalose , Obesity/etiology , Diet , Drosophila , Fertility , Glucose , Glycogen
2.
Sci Rep ; 13(1): 12331, 2023 07 30.
Article in English | MEDLINE | ID: mdl-37518578

ABSTRACT

Temperature influences male fertility across organisms; however, how suboptimal temperatures affect adult spermatogenesis remains understudied. In a recent study on Drosophila melanogaster oogenesis, we observed a drastic reduction in the fertility of adult males exposed to warm temperature (29 °C). Here, we show that males become infertile at 29 °C because of low sperm abundance and quality. The low sperm abundance at 29 °C does not stem from reduced germline stem cell or spermatid numbers, as those numbers remain comparable between 29 °C and control 25 °C. Notably, males at cold 18 °C and 29 °C had similarly increased frequencies of spermatid elongation and individualization defects which, considering the high sperm abundance and male fertility measured at 18 °C, indicate that spermatogenesis has a high tolerance for elongation and individualization defects. Interestingly, the abundance of sperm at 29 °C decreases abruptly and with no evidence of apoptosis as they transition into the seminal vesicle near the end of spermatogenesis, pointing to sperm elimination through an unknown mechanism. Finally, sperm from males at 29 °C fertilize eggs less efficiently and do not support embryos past the first stage of embryogenesis, indicating that poor sperm quality is an additional cause of male infertility at 29 °C.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Animals , Male , Temperature , Semen , Spermatozoa , Spermatogenesis , Fertility
3.
Development ; 149(5)2022 03 01.
Article in English | MEDLINE | ID: mdl-35156684

ABSTRACT

Despite their medical and economic relevance, it remains largely unknown how suboptimal temperatures affect adult insect reproduction. Here, we report an in-depth analysis of how chronic adult exposure to suboptimal temperatures affects oogenesis using the model insect Drosophila melanogaster. In adult females maintained at 18°C (cold) or 29°C (warm), relative to females at the 25°C control temperature, egg production was reduced through distinct cellular mechanisms. Chronic 18°C exposure improved germline stem cell maintenance, survival of early germline cysts and oocyte quality, but reduced follicle growth with no obvious effect on vitellogenesis. By contrast, in females at 29°C, germline stem cell numbers and follicle growth were similar to those at 25°C, while early germline cyst death and degeneration of vitellogenic follicles were markedly increased and oocyte quality plummeted over time. Finally, we also show that these effects are largely independent of diet, male factors or canonical temperature sensors. These findings are relevant not only to cold-blooded organisms, which have limited thermoregulation, but also potentially to warm-blooded organisms, which are susceptible to hypothermia, heatstroke and fever.


Subject(s)
Cell Lineage/physiology , Drosophila melanogaster/physiology , Germ Cells/physiology , Oogenesis/physiology , Stem Cells/physiology , Animals , Cold Temperature , Female , Gene Expression Regulation, Developmental/physiology , Male , Oocytes/physiology , Ovarian Follicle/physiology , Ovary/physiology , Signal Transduction/physiology , Vitellogenesis/physiology
4.
Development ; 148(5)2021 03 09.
Article in English | MEDLINE | ID: mdl-33547134

ABSTRACT

The conserved nuclear receptor superfamily has crucial roles in many processes, including reproduction. Nuclear receptors with known roles in oogenesis have been studied mostly in the context of their ovary-intrinsic requirement. Recent studies in Drosophila, however, have begun to reveal new roles of nuclear receptor signaling in peripheral tissues in controlling reproduction. Here, we identified Hormone receptor 4 (Hr4) as an oogenesis regulator required in the ovary and muscles. Global Hr4 knockdown leads to increased germline stem cell (GSC) loss, reduced GSC proliferation, early germline cyst death, slowed follicle growth and vitellogenic follicle degeneration. Tissue-specific knockdown experiments uncovered ovary-intrinsic and peripheral tissue requirements for Hr4 In the ovary, Hr4 is required in the niche for GSC proliferation and in the germline for GSC maintenance. Hr4 functions in muscles to promote GSC maintenance and follicle growth. The specific tissues that require Hr4 for survival of early germline cysts and vitellogenic follicles remain unidentified. These results add to the few examples of muscles controlling gametogenesis and expand our understanding of the complexity of nuclear receptor regulation of various aspects of oogenesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Muscles/metabolism , Oogenesis/genetics , Ovary/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Animals, Genetically Modified/metabolism , Cell Proliferation , Drosophila/growth & development , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Female , Germ Cells/cytology , Germ Cells/metabolism , Muscles/cytology , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovary/cytology , Ovum/growth & development , Ovum/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Receptors, Cytoplasmic and Nuclear/genetics
5.
PLoS One ; 15(12): e0243756, 2020.
Article in English | MEDLINE | ID: mdl-33307547

ABSTRACT

Reproduction is highly sensitive to changes in physiology and the external environment. Neuropeptides are evolutionarily conserved signaling molecules that regulate multiple physiological processes. However, the potential reproductive roles of many neuropeptide signaling pathways remain underexplored. Here, we describe the results of RNAi-based screens in Drosophila melanogaster to identify neuropeptides/neuropeptide receptors with potential roles in oogenesis. The screen read-outs were either the number of eggs laid per female per day over time or fluorescence microscopy analysis of dissected ovaries. We found that the orphan neuropeptide receptor encoded by moody (homologous to mammalian melatonin receptors) is likely required in somatic cells for normal egg production and proper germline stem cell maintenance. However, the egg laying screens had low signal-to-noise ratio and did not lead to the identification of additional candidates. Thus, although egg count assays might be useful for large-scale screens to identify oogenesis regulators that result in dramatic changes in oogenesis, more labor-intensive microscopy-based screen are better applicable for identifying new physiological regulators of oogenesis with more subtle phenotypes.


Subject(s)
Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Oogonial Stem Cells/cytology , RNA Interference , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Animals , Drosophila melanogaster/genetics , Female , Neuropeptides/metabolism , Oogenesis , Oogonial Stem Cells/metabolism
6.
G3 (Bethesda) ; 10(12): 4625-4635, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33087412

ABSTRACT

The physiology of organisms depends on inter-organ communication in response to changes in the environment. Nuclear receptors are broadly expressed transcription factors that respond to circulating molecules to control many biological processes, including immunity, detoxification, and reproduction. Although the tissue-intrinsic roles of nuclear receptors in reproduction have been extensively studied, there is increasing evidence that nuclear receptor signaling in peripheral tissues can also influence oogenesis. We previously showed that the Drosophila nuclear receptor Seven up (Svp) is required in the adult fat body to regulate distinct steps of oogenesis; however, the relevant downstream targets of Svp remain unknown. Here, we took an RNA sequencing approach to identify candidate Svp targets specifically in the adult female fat body that might mediate this response. svp knockdown in the adult female fat body significantly downregulated immune genes involved in the first line of pathogen defense, suggesting a role for Svp in stimulating early immunity. In addition, we found that Svp transcriptionally regulates genes involved in each step of the xenobiotic detoxification response. Based on these findings, we propose a testable model in which Svp functions in the adult female fat body to stimulate early defense against pathogens and facilitate detoxification as part of its mechanisms to promote oogenesis.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila Proteins/genetics , Fat Body , Female , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors , Xenobiotics
7.
G3 (Bethesda) ; 10(11): 4147-4158, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32917721

ABSTRACT

Precise genetic manipulation of specific cell types or tissues to pinpoint gene function requirement is a critical step in studies aimed at unraveling the intricacies of organismal physiology. Drosophila researchers heavily rely on the UAS/Gal4/Gal80 system for tissue-specific manipulations; however, it is often unclear whether the reported Gal4 expression patterns are indeed specific to the tissue of interest such that experimental results are not confounded by secondary sites of Gal4 expression. Here, we surveyed the expression patterns of commonly used Gal4 drivers in adult Drosophila female tissues under optimal conditions and found that multiple drivers have unreported secondary sites of expression beyond their published cell type/tissue expression pattern. These results underscore the importance of thoroughly characterizing Gal4 tools as part of a rigorous experimental design that avoids potential misinterpretation of results as we strive for understanding how the function of a specific gene/pathway in one tissue contributes to whole-body physiology.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Female , Genetic Techniques , Transcription Factors/genetics
8.
Development ; 147(11)2020 06 14.
Article in English | MEDLINE | ID: mdl-32540896

ABSTRACT

Developmental biologists have frequently pushed the frontiers of modern biomedical research. From the discovery and characterization of novel signal transduction pathways to exploring the molecular underpinnings of genetic inheritance, transcription, the cell cycle, cell death and stem cell biology, studies of metazoan development have historically opened new fields of study and consistently revealed previously unforeseen avenues of clinical therapies. From this perspective, it is not surprising that our community is now an integral part of the current renaissance in metabolic research. Amidst the global rise in metabolic syndrome, the discovery of novel signaling roles for metabolites, and the increasing links between altered metabolism and many human diseases, we as developmental biologists can contribute skills and expertise that are uniquely suited for investigating the mechanisms underpinning human metabolic health and disease. Here, we summarize the opportunities and challenges that our community faces, and discuss how developmental biologists can make unique and valuable contributions to the field of metabolism and physiology.


Subject(s)
Developmental Biology , Metabolic Diseases/metabolism , Animals , Drosophila/growth & development , Drosophila/metabolism , Humans , Metabolic Diseases/pathology , Neoplasms/metabolism , Neoplasms/pathology , Oogenesis , Sea Urchins/growth & development , Sea Urchins/metabolism , Signal Transduction
9.
Genetics ; 213(1): 9-26, 2019 09.
Article in English | MEDLINE | ID: mdl-31488592

ABSTRACT

The long-term survival of any multicellular species depends on the success of its germline in producing high-quality gametes and maximizing survival of the offspring. Studies in Drosophila melanogaster have led our growing understanding of how germline stem cell (GSC) lineages maintain their function and adjust their behavior according to varying environmental and/or physiological conditions. This review compares and contrasts the local regulation of GSCs by their specialized microenvironments, or niches; discusses how diet and diet-dependent factors, mating, and microorganisms modulate GSCs and their developing progeny; and briefly describes the tie between physiology and development during the larval phase of the germline cycle. Finally, it concludes with broad comparisons with other organisms and some future directions for further investigation.


Subject(s)
Drosophila melanogaster/genetics , Gametogenesis , Gene Expression Regulation, Developmental , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Drosophila melanogaster/growth & development , Gene-Environment Interaction , Stem Cell Niche
10.
Dev Biol ; 456(2): 179-189, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31470019

ABSTRACT

Reproduction is intimately linked to the physiology of an organism. Nuclear receptors are widely expressed transcription factors that mediate the effects of many circulating molecules on physiology and reproduction. While multiple studies have focused on the roles of nuclear receptors intrinsically in the ovary, it remains largely unknown how the actions of nuclear receptors in peripheral tissues influence oogenesis. We identified the nuclear receptor encoded by svp as a novel regulator of oogenesis in adult Drosophila. Global somatic knockdown of svp reduces egg production by increasing GSC loss, death of early germline cysts, and degeneration of vitellogenic follicles. Tissue-specific knockdown experiments revealed that svp remotely controls these different steps of oogenesis through separate mechanisms involving distinct tissues. Specifically, adipocyte-specific svp knockdown impairs GSC maintenance and early germline cyst survival, whereas oenocyte-specific svp knockdown increases the death of vitellogenic follicles without any effects on GSCs or early cysts. These results illustrate that nuclear receptors can control reproduction through a variety of mechanisms involving peripheral tissues.


Subject(s)
Adipocytes/metabolism , DNA-Binding Proteins/metabolism , Oogenesis/physiology , Receptors, Steroid/metabolism , Adipocytes/physiology , Animals , DNA-Binding Proteins/physiology , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Germ Cells/metabolism , Ovary/metabolism , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid/physiology , Signal Transduction/physiology , Transcription Factors
11.
Genetics ; 209(4): 1155-1166, 2018 08.
Article in English | MEDLINE | ID: mdl-29884747

ABSTRACT

Stem cells reside in specialized niches and are regulated by a variety of physiological inputs. Adipocytes influence whole-body physiology and stem cell lineages; however, the molecular mechanisms linking adipocytes to stem cells are poorly understood. Here, we report that collagen IV produced in adipocytes is transported to the ovary to maintain proper germline stem cell (GSC) number in adult Drosophila females. Adipocyte-derived collagen IV acts through ß-integrin signaling to maintain normal levels of E-cadherin at the niche, thereby ensuring proper adhesion to GSCs. These findings demonstrate that extracellular matrix components produced in adipocytes can be transported to and incorporated into an established adult tissue to influence stem cell number.


Subject(s)
Adipocytes/metabolism , Collagen Type IV/metabolism , Drosophila Proteins/metabolism , Drosophila/physiology , Extracellular Matrix Proteins/metabolism , Ovary/cytology , Animals , Cadherins/metabolism , Cell Count , Cell Lineage , Cells, Cultured , Drosophila/cytology , Drosophila/metabolism , Female , Focal Adhesion Kinase 1/metabolism , Integrin beta Chains/metabolism , Ovary/metabolism , Protein Transport , Signal Transduction , Stem Cell Niche , Stem Cells/cytology , Stem Cells/metabolism
12.
Dev Biol ; 440(1): 31-39, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29729259

ABSTRACT

Tissue-specific stem cells are tied to the nutritional and physiological environment of adult organisms. Adipocytes have key endocrine and nutrient-sensing roles and have emerged as major players in relaying dietary information to regulate other organs. For example, previous studies in Drosophila melanogaster revealed that amino acid sensing as well as diet-dependent metabolic pathways function in adipocytes to influence the maintenance of female germline stem cells (GSCs). How nutrient-sensing pathways acting within adipocytes influence adult stem cell lineages, however, is just beginning to be elucidated. Here, we report that insulin/insulin-like growth factor signaling in adipocytes promotes GSC maintenance, early germline cyst survival, and vitellogenesis. Further, adipocytes use distinct mechanisms downstream of insulin receptor activation to control these aspects of oogenesis, all of which are independent of FOXO. We find that GSC maintenance is modulated by Akt1 through GSK-3ß, early germline cyst survival is downstream of adipocyte Akt1 but independent of GSK-3ß, and vitellogenesis is regulated through an Akt1-independent pathway in adipocytes. These results indicate that, in addition to employing different types of nutrient sensing, adipocytes can use distinct axes of a single nutrient-sensing pathway to regulate multiple stages of the GSC lineage in the ovary.


Subject(s)
Adipocytes/physiology , Adult Germline Stem Cells/physiology , Glycogen Synthase Kinase 3 beta/metabolism , Adipocytes/metabolism , Adult Germline Stem Cells/metabolism , Animals , Cell Count , Cell Proliferation , Drosophila/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Female , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/physiology , Germ Cells/cytology , Glycogen Synthase Kinase 3 beta/physiology , Insulin/metabolism , Male , Oogenesis/physiology , Ovary/cytology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Stem Cells/cytology
13.
Curr Stem Cell Rep ; 3(1): 9-18, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28458991

ABSTRACT

PURPOSE OF REVIEW: Stem cells respond to local paracrine signals; more recently, however, systemic hormones have also emerged as key regulators of stem cells. This review explores the role of steroid hormones in stem cells, using the Drosophila germline stem cell as a centerpiece for discussion. RECENT FINDINGS: Stem cells sense and respond directly and indirectly to steroid hormones, which regulate diverse sets of target genes via interactions with nuclear hormone receptors. Hormone-regulated networks likely integrate the actions of multiple systemic signals to adjust the activity of stem cell lineages in response to changes in physiological status. SUMMARY: Hormones are inextricably linked to animal physiology, and can control stem cells and their local niches. Elucidating the molecular mechanisms of hormone signaling in stem cells is essential for our understanding of the fundamental underpinnings of stem cell biology, and for informing new therapeutic interventions against cancers or for regenerative medicine.

14.
Genetics ; 206(2): 953-971, 2017 06.
Article in English | MEDLINE | ID: mdl-28396508

ABSTRACT

Nutrients affect adult stem cells through complex mechanisms involving multiple organs. Adipocytes are highly sensitive to diet and have key metabolic roles, and obesity increases the risk for many cancers. How diet-regulated adipocyte metabolic pathways influence normal stem cell lineages, however, remains unclear. Drosophila melanogaster has highly conserved adipocyte metabolism and a well-characterized female germline stem cell (GSC) lineage response to diet. Here, we conducted an isobaric tags for relative and absolute quantification (iTRAQ) proteomic analysis to identify diet-regulated adipocyte metabolic pathways that control the female GSC lineage. On a rich (relative to poor) diet, adipocyte Hexokinase-C and metabolic enzymes involved in pyruvate/acetyl-CoA production are upregulated, promoting a shift of glucose metabolism toward macromolecule biosynthesis. Adipocyte-specific knockdown shows that these enzymes support early GSC progeny survival. Further, enzymes catalyzing fatty acid oxidation and phosphatidylethanolamine synthesis in adipocytes promote GSC maintenance, whereas lipid and iron transport from adipocytes controls vitellogenesis and GSC number, respectively. These results show a functional relationship between specific metabolic pathways in adipocytes and distinct processes in the GSC lineage, suggesting the adipocyte metabolism-stem cell link as an important area of investigation in other stem cell systems.


Subject(s)
Cell Lineage/genetics , Germ Cells/growth & development , Metabolic Networks and Pathways/genetics , Proteomics , Adipocytes/metabolism , Animals , Diet , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Fatty Acids/genetics , Fatty Acids/metabolism , Female , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Hexokinase/biosynthesis , Hexokinase/genetics , Oogonial Stem Cells/metabolism , Phosphatidylethanolamines/biosynthesis , Phosphatidylethanolamines/genetics , Vitellogenesis/genetics
15.
Results Probl Cell Differ ; 59: 67-99, 2017.
Article in English | MEDLINE | ID: mdl-28247046

ABSTRACT

Tight coupling of reproduction to environmental factors and physiological status is key to long-term species survival. In particular, highly conserved pathways modulate germline stem cell lineages according to nutrient availability. This chapter focuses on recent in vivo studies in genetic model organisms that shed light on how diet-dependent signals control the proliferation, maintenance, and survival of adult germline stem cells and their progeny. These signaling pathways can operate intrinsically in the germ line, modulate the niche, or act through intermediate organs to influence stem cells and their differentiating progeny. In addition to illustrating the extent of dietary regulation of reproduction, findings from these studies have implications for fertility during aging or disease states.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Diet , Germ Cells/cytology , Stress, Physiological/physiology , Animals , Humans
16.
Gene Expr Patterns ; 23-24: 13-21, 2017 01.
Article in English | MEDLINE | ID: mdl-28093350

ABSTRACT

The effect of diet on reproduction is well documented in a large number of organisms; however, much remains to be learned about the molecular mechanisms underlying this connection. The Drosophila ovary has a well described, fast and largely reversible response to diet. Ovarian stem cells and their progeny proliferate and grow faster on a yeast-rich diet than on a yeast-free (poor) diet, and death of early germline cysts, degeneration of early vitellogenic follicles and partial block in ovulation further contribute to the ∼60-fold decrease in egg laying observed on a poor diet. Multiple diet-dependent factors, including insulin-like peptides, the steroid ecdysone, the nutrient sensor Target of Rapamycin, AMP-dependent kinase, and adipocyte factors mediate this complex response. Here, we describe the results of a visual screen using a collection of green fluorescent protein (GFP) protein trap lines to identify additional factors potentially involved in this response. In each GFP protein trap line, an artificial GFP exon is fused in frame to an endogenous protein, such that the GFP fusion pattern parallels the levels and subcellular localization of the corresponding native protein. We identified 53 GFP-tagged proteins that exhibit changes in levels and/or subcellular localization in the ovary at 12-16 hours after switching females from rich to poor diets, suggesting them as potential candidates for future functional studies.


Subject(s)
Drosophila Proteins/analysis , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Animals , Diet , Female , Green Fluorescent Proteins/analysis , Oogenesis , Ovary/chemistry , Ovary/metabolism , Ovum/chemistry , Recombinant Fusion Proteins/analysis , Yeasts
17.
Dev Biol ; 420(1): 90-99, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27729213

ABSTRACT

Multiple aspects of organismal physiology influence the number and activity of stem cells and their progeny, including nutritional status. Previous studies demonstrated that Drosophila germline stem cells (GSCs), follicle stem cells (FSCs), and their progeny sense and respond to diet via complex mechanisms involving many systemic and local signals. AMP-activated protein kinase, or AMPK, is a highly conserved regulator of energy homeostasis known to be activated under low cellular energy conditions; however, its role in the ovarian response to diet has not been investigated. Here, we describe nutrient-dependent and -independent requirements for AMPK in Drosophila oogenesis. We found that AMPK is cell autonomously required for the slow down in GSC and follicle cell proliferation that occurs on a poor diet. Similarly, AMPK activity is necessary in the germline for the degeneration of vitellogenic stages in response to nutrient deprivation. In contrast, AMPK activity is not required within the germline to modulate its growth. Instead, AMPK acts in follicle cells to negatively regulate their growth and proliferation, thereby indirectly limiting the size of the underlying germline cyst within developing follicles. Paradoxically, AMPK is required for GSC maintenance in well-fed flies (when AMPK activity is presumably at its lowest), suggesting potentially important roles for basal AMPK activity in specific cell types. Finally, we identified a nutrient-independent, developmental role for AMPK in cyst encapsulation by follicle cells. These results uncover specific AMPK requirements in multiple cell types in the ovary and suggest that AMPK can function outside of its canonical nutrient-sensing role in specific developmental contexts.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diet , Drosophila melanogaster/metabolism , Oogenesis , Animals , Cell Proliferation , Cell Size , Down-Regulation , Endoreduplication , Feeding Behavior , Female , Germ Cells/cytology , Mitosis , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Vitellogenins/metabolism
18.
G3 (Bethesda) ; 6(8): 2629-42, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27226164

ABSTRACT

Multiple aspects of Drosophila oogenesis, including germline stem cell activity, germ cell differentiation, and follicle survival, are regulated by the steroid hormone ecdysone. While the transcriptional targets of ecdysone signaling during development have been studied extensively, targets in the ovary remain largely unknown. Early studies of salivary gland polytene chromosomes led to a model in which ecdysone stimulates a hierarchical transcriptional cascade, wherein a core group of ecdysone-sensitive transcription factors induce tissue-specific responses by activating secondary branches of transcriptional targets. More recently, genome-wide approaches have identified hundreds of putative ecdysone-responsive targets. Determining whether these putative targets represent bona fide targets in vivo, however, requires that they be tested via traditional mutant analysis in a cell-type specific fashion. To investigate the molecular mechanisms whereby ecdysone signaling regulates oogenesis, we used genetic mosaic analysis to screen putative ecdysone-responsive genes for novel roles in the control of the earliest steps of oogenesis. We identified a cohort of genes required for stem cell maintenance, stem and progenitor cell proliferation, and follicle encapsulation, growth, and survival. These genes encode transcription factors, chromatin modulators, and factors required for RNA transport, stability, and ribosome biogenesis, suggesting that ecdysone might control a wide range of molecular processes during oogenesis. Our results suggest that, although ecdysone target genes are known to have cell type-specific roles, many ecdysone response genes that control larval or pupal cell types at developmental transitions are used reiteratively in the adult ovary. These results provide novel insights into the molecular mechanisms by which ecdysone signaling controls oogenesis, laying new ground for future studies.


Subject(s)
Drosophila melanogaster/genetics , Ecdysone/metabolism , Gene Expression Regulation, Developmental , Oogenesis/genetics , Ovary/physiology , Animals , Animals, Genetically Modified , Cell Lineage , Cell Survival/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Ecdysone/pharmacology , Female , Gene Expression Regulation, Developmental/drug effects , Genetic Techniques , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Larva/drug effects , Larva/genetics , Mosaicism , Mutation , Oogenesis/drug effects , Ovary/cytology , Pupa/genetics , Receptors, Steroid/genetics , Stem Cells/physiology
19.
Methods Mol Biol ; 1328: 57-72, 2015.
Article in English | MEDLINE | ID: mdl-26324429

ABSTRACT

Genetic mosaic analyses represent an invaluable approach for the study of stem cell lineages in the Drosophila ovary. The generation of readily identifiable, homozygous mutant cells in the context of wild-type ovarian tissues within intact organisms allows the pinpointing of cellular requirements for gene function, which is particularly important for understanding the physiological control of stem cells and their progeny. Here, we provide a step-by-step guide to the generation and analysis of genetically mosaic ovaries using flippase (FLP)/FLP recognition target (FRT)-mediated recombination in adult Drosophila melanogaster, with a focus on the processes of oogenesis that are controlled by diet-dependent factors.


Subject(s)
DNA Nucleotidyltransferases/genetics , Drosophila melanogaster/genetics , Molecular Biology/methods , Mosaicism , Animals , Cell Lineage/genetics , Female , Ovary/growth & development , Stem Cells/cytology
20.
Nat Cell Biol ; 17(6): 707-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26022916

ABSTRACT

Control of stem cell activity is essential for accurate regeneration. Pathogen- or chemical-induced intestinal damage is now shown to recruit haemocytes expressing bone morphogenetic protein signals that stimulate proliferation of intestinal stem cells and subsequently induce their quiescence, in conjunction with muscle-derived bone morphogenetic proteins. A temporal switch in expression of Type I receptors enables this two-phase response.


Subject(s)
Drosophila melanogaster/cytology , Hemocytes/cytology , Intestinal Mucosa/cytology , Stem Cells/cytology , Tight Junctions/immunology , Animals
SELECTION OF CITATIONS
SEARCH DETAIL