Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38585877

ABSTRACT

Measurements of Drosophila fecundity are used in a wide variety of studies, such as investigations of stem cell biology, nutrition, behavior, and toxicology. In addition, because fecundity assays are performed on live flies, they are suitable for longitudinal studies such as investigations of aging or prolonged chemical exposure. However, standard Drosophila fecundity assays have been difficult to perform in a high-throughput manner because experimental factors such as the physiological state of the flies and environmental cues must be carefully controlled to achieve consistent results. In addition, exposing flies to a large number of different experimental conditions (such as chemical additives in the diet) and manually counting the number of eggs laid to determine the impact on fecundity is time-consuming. We have overcome these challenges by combining a new multiwell fly culture strategy with a novel 3D-printed fly transfer device to rapidly and accurately transfer flies from one plate to another; the RoboCam, a low-cost, custom built robotic camera to capture images of the wells automatically; and an image segmentation pipeline to automatically identify and quantify eggs. We show that this method is compatible with robust and consistent egg laying throughout the assay period; and demonstrate that the automated pipeline for quantifying fecundity is very accurate (r2 = 0.98 for the correlation between the automated egg counts and the ground truth) In addition, we show that this method can be used to efficiently detect the effects on fecundity induced by dietary exposure to chemicals. Taken together, this strategy substantially increases the efficiency and reproducibility of high throughput egg laying assays that require exposing flies to multiple different media conditions.

2.
EMBO Rep ; 24(10): e56475, 2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37603128

ABSTRACT

Drosophila ovarian germline stem cells (GSCs) are a powerful model for stem cell research. In this study, we use single-cell RNA sequencing (scRNA-seq), an RNAi screen and bioinformatic analysis, to identify genes involved in germ cell differentiation, including 34 genes with upregulated expression during early germ cell development and 19 genes that may regulate germ cell differentiation. Among these, a gene we have named eggplant (eggpl) is highly expressed in GSCs and downregulated in early daughter cells. RNAi knockdown of eggpl causes germ cell proliferation and differentiation defects. In flies fed a rich yeast diet, the expression of eggpl is significantly lower and knockdown or knockout of eggpl phenocopies a rich diet. In addition, eggpl knockdown suppresses the reduction in germ cell proliferation caused by inhibition of the insulin effector PI3K. These findings suggest that downregulation of eggpl links nutritional status to germ cell proliferation and differentiation. Collectively, this study provides new insights into the signaling networks that regulate early germ cell development and identifies eggpl as a key player in this process.


Subject(s)
Drosophila Proteins , Solanum melongena , Animals , Drosophila/genetics , Solanum melongena/genetics , Solanum melongena/metabolism , Drosophila Proteins/metabolism , Cell Differentiation/genetics , Germ Cells/metabolism , Sequence Analysis, RNA , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism
3.
Nat Commun ; 14(1): 3745, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37353491

ABSTRACT

Intracellular pH dynamics is increasingly recognized to regulate myriad cell behaviors. We report a finding that intracellular pH dynamics also regulates adult stem cell lineage specification. We identify an intracellular pH gradient in mouse small intestinal crypts, lowest in crypt stem cells and increasing along the crypt column. Disrupting this gradient by inhibiting H+ efflux by Na+/H+ exchanger 1 abolishes crypt budding and blocks differentiation of Paneth cells, which are rescued with exogenous WNT. Using single-cell RNA sequencing and lineage tracing we demonstrate that intracellular pH dynamics acts downstream of ATOH1, with increased pH promoting differentiation toward the secretory lineage. Our findings indicate that an increase in pH is required for the lineage specification that contributes to crypt maintenance, establishing a role for intracellular pH dynamics in cell fate decisions within an adult stem cell lineage.


Subject(s)
Intestines , Stem Cells , Mice , Animals , Cell Lineage , Cell Differentiation/physiology , Hydrogen-Ion Concentration , Intestinal Mucosa
4.
Cell Stem Cell ; 30(2): 188-206.e6, 2023 02 02.
Article in English | MEDLINE | ID: mdl-36640764

ABSTRACT

A central factor in the maintenance of tissue integrity is the response of stem cells to variations in the levels of niche signals. In the gut, intestinal stem cells (ISCs) depend on Wnt ligands for self-renewal and proliferation. Transient increases in Wnt signaling promote regeneration after injury or in inflammatory bowel diseases, whereas constitutive activation of this pathway leads to colorectal cancer. Here, we report that Discs large 1 (Dlg1), although dispensable for polarity and cellular turnover during intestinal homeostasis, is required for ISC survival in the context of increased Wnt signaling. RNA sequencing (RNA-seq) and genetic mouse models demonstrated that DLG1 regulates the cellular response to increased canonical Wnt ligands. This occurs via the transcriptional regulation of Arhgap31, a GTPase-activating protein that deactivates CDC42, an effector of the non-canonical Wnt pathway. These findings reveal a DLG1-ARHGAP31-CDC42 axis that is essential for the ISC response to increased niche Wnt signaling.


Subject(s)
Intestinal Mucosa , Wnt Signaling Pathway , Animals , Mice , Cell Proliferation , GTPase-Activating Proteins/metabolism , Intestinal Mucosa/metabolism , Intestines , Stem Cell Niche , Stem Cells , Wnt Signaling Pathway/genetics
5.
Methods Mol Biol ; 2626: 323-333, 2023.
Article in English | MEDLINE | ID: mdl-36715913

ABSTRACT

The production of eggs in the Drosophila ovary requires complex interactions between multiple cell types that coexist within the same solid tissue. This cellular heterogeneity makes the ovary a rich subject of study, but also makes it challenging to identify transcriptional differences between individual cell types using methods such as bulk RNA sequencing. The development of single-cell RNA sequencing (scRNA-seq) techniques addresses this limitation by providing an avenue to profile genetic and functional heterogeneity at a cellular resolution. Here, we describe the isolation and preparation of the Drosophila ovary for scRNA-seq. This protocol emphasizes a short preparation time, high cell viability, prevention of RNA-degradation, and reduction of technical variation to achieve highly reproducible single-cell profiles.


Subject(s)
Drosophila , Single-Cell Analysis , Animals , Female , Drosophila/genetics , Single-Cell Analysis/methods , Sequence Analysis, RNA/methods , Ovary/metabolism , Base Sequence , RNA/genetics , RNA/metabolism , Gene Expression Profiling/methods
6.
Science ; 375(6584): eabk2432, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35239393

ABSTRACT

For more than 100 years, the fruit fly Drosophila melanogaster has been one of the most studied model organisms. Here, we present a single-cell atlas of the adult fly, Tabula Drosophilae, that includes 580,000 nuclei from 15 individually dissected sexed tissues as well as the entire head and body, annotated to >250 distinct cell types. We provide an in-depth analysis of cell type-related gene signatures and transcription factor markers, as well as sexual dimorphism, across the whole animal. Analysis of common cell types between tissues, such as blood and muscle cells, reveals rare cell types and tissue-specific subtypes. This atlas provides a valuable resource for the Drosophila community and serves as a reference to study genetic perturbations and disease models at single-cell resolution.


Subject(s)
Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Transcriptome , Animals , Cell Nucleus/metabolism , Databases, Genetic , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Female , Gene Expression Regulation , Gene Regulatory Networks , Genes, Insect , Male , RNA-Seq , Sex Characteristics , Single-Cell Analysis , Transcription Factors/genetics
8.
Development ; 148(22)2021 11 15.
Article in English | MEDLINE | ID: mdl-35020878

ABSTRACT

A major goal in the study of adult stem cells is to understand how cell fates are specified at the proper time and place to facilitate tissue homeostasis. Here, we found that an E2 ubiquitin ligase, Bendless (Ben), has multiple roles in the Drosophila ovarian epithelial follicle stem cell (FSC) lineage. First, Ben is part of the JNK signaling pathway, and we found that it, as well as other JNK pathway genes, are essential for differentiation of FSC daughter cells. Our data suggest that JNK signaling promotes differentiation by suppressing the activation of the EGFR effector, ERK. Also, we found that loss of ben, but not the JNK kinase hemipterous, resulted in an upregulation of hedgehog signaling, increased proliferation and increased niche competition. Lastly, we demonstrate that the hypercompetition phenotype caused by loss of ben is suppressed by decreasing the rate of proliferation or knockdown of the hedgehog pathway effector, Smoothened (Smo). Taken together, our findings reveal a new layer of regulation in which a single gene influences cell signaling at multiple stages of differentiation in the early FSC lineage.


Subject(s)
Drosophila Proteins/genetics , ErbB Receptors/genetics , Hedgehog Proteins/genetics , Ovarian Follicle/growth & development , Receptors, Invertebrate Peptide/genetics , Smoothened Receptor/genetics , Ubiquitin-Conjugating Enzymes/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Proliferation/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Epithelial Cells/cytology , Female , MAP Kinase Signaling System/genetics , Ovarian Follicle/metabolism , Stem Cell Niche/genetics , Stem Cells/cytology
9.
Nat Commun ; 11(1): 5628, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159074

ABSTRACT

The Drosophila ovary is a widely used model for germ cell and somatic tissue biology. Here we use single-cell RNA-sequencing (scRNA-seq) to build a comprehensive cell atlas of the adult Drosophila ovary that contains transcriptional profiles for every major cell type in the ovary, including the germline stem cells and their niche cells, follicle stem cells, and previously undescribed subpopulations of escort cells. In addition, we identify Gal4 lines with specific expression patterns and perform lineage tracing of subpopulations of escort cells and follicle cells. We discover that a distinct subpopulation of escort cells is able to convert to follicle stem cells in response to starvation or upon genetic manipulation, including knockdown of escargot, or overactivation of mTor or Toll signalling.


Subject(s)
Drosophila/cytology , Ovary/cytology , Animals , Cell Lineage , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Female , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Ovary/metabolism , Single-Cell Analysis
10.
Curr Opin Insect Sci ; 37: 39-48, 2020 02.
Article in English | MEDLINE | ID: mdl-32087562

ABSTRACT

The follicle stem cell (FSC) lineage in the Drosophila ovary is a highly informative model of in vivo epithelial stem cell biology. Studies over the past 30 years have identified roles for every major signaling pathway in the early FSC lineage. These pathways regulate a wide variety of cell behaviors, including self-renewal, proliferation, survival and differentiation. Studies of cell signaling in the follicle epithelium have provided new insights into how these cell behaviors are coordinated within an epithelial stem cell lineage and how signaling pathways interact with each other in the native, in vivo context of a living tissue. Here, we review these studies, with a particular focus on how these pathways specify differences between the FSCs and their daughter cells. We also describe common themes that have emerged from these studies, and highlight new research directions that have been made possible by the detailed understanding of the follicle epithelium.


Subject(s)
Ovarian Follicle/cytology , Signal Transduction , Stem Cells/metabolism , Animals , Drosophila/growth & development , Female , Ovarian Follicle/metabolism , Stem Cell Niche , Stem Cells/cytology
11.
Elife ; 82019 12 18.
Article in English | MEDLINE | ID: mdl-31850843

ABSTRACT

The follicle stem cells (FSCs) in the Drosophila ovary are an important experimental model for the study of epithelial stem cell biology. Although decades of research support the conclusion that there are two FSCs per ovariole, a recent study used a novel clonal marking system to conclude that there are 15-16 FSCs per ovariole. We performed clonal analysis using both this novel clonal marking system and standard clonal marking systems, and identified several problems that may have contributed to the overestimate of FSC number. In addition, we developed new methods for accurately measuring clone size, and found that FSC clones produce, on average, half of the follicle cells in each ovariole. Our findings provide strong independent support for the conclusion that there are typically two active FSCs per ovariole, though they are consistent with up to four FSCs per germarium.


Subject(s)
Drosophila melanogaster/cytology , Epithelial Cells/cytology , Ovarian Follicle/cytology , Stem Cells/cytology , Stem Cells/physiology , Animals , Female , Ovary
12.
Dev Biol ; 452(2): 127-133, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31071312

ABSTRACT

Understanding how cell fate decisions are regulated is a central question in stem cell biology. Recent studies have demonstrated that intracellular pH (pHi) dynamics contribute to this process. Indeed, the pHi of cells within a tissue is not simply a consequence of chemical reactions in the cytoplasm and other cellular activity, but is actively maintained at a specific setpoint in each cell type. We found previously that the pHi of cells in the follicle stem cell (FSC) lineage in the Drosophila ovary increases progressively during differentiation from an average of 6.8 in the FSCs, to 7.0 in newly produced daughter cells, to 7.3 in more differentiated cells. Two major regulators of pHi in this lineage are Drosophila sodium-proton exchanger 2 (dNhe2) and a previously uncharacterized gene, CG8177, that is homologous to mammalian anion exchanger 2 (AE2). Based on this homology, we named the gene anion exchanger 2 (ae2). Here, we generated null alleles of ae2 and found that homozygous mutant flies are viable but have severe defects in ovary development and adult oogenesis. Specifically, we find that ae2 null flies have smaller ovaries, reduced fertility, and impaired follicle formation. In addition, we find that the follicle formation defect can be suppressed by a decrease in dNhe2 copy number and enhanced by the overexpression of dNhe2, suggesting that this phenotype is due to the dysregulation of pHi. These findings support the emerging idea that pHi dynamics regulate cell fate decisions and our studies provide new genetic tools to investigate the mechanisms by which this occurs.


Subject(s)
Antiporters/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Oogenesis , Ovary/embryology , Ovary/metabolism , Animals , Drosophila melanogaster/genetics , Epistasis, Genetic , Female , Fertility , Mutation/genetics , Organ Size , Ovarian Follicle/embryology , RNA Interference , Sequence Homology, Amino Acid
13.
Development ; 145(23)2018 12 05.
Article in English | MEDLINE | ID: mdl-30389852

ABSTRACT

Adult stem cell niche boundaries must be precisely maintained to facilitate the segregation of stem cell and daughter cell fates. However, the mechanisms that govern this process in epithelial tissues are not fully understood. In this study, we investigated the relationship between two signals, Wnt and EGFR, that are necessary for self-renewal of the epithelial follicle stem cells (FSCs) in the Drosophila ovary, but must be downregulated in cells that have exited the niche to allow for differentiation. We found that Wingless produced by inner germarial sheath (IGS) cells acts over a short distance to activate Wnt signaling in FSCs, and that movement across the FSC niche boundary is limited. In addition, we show that Wnt signaling functions genetically upstream of EGFR signaling by activating the expression of the EGFR ligand, Spitz, and that constitutive activation of EGFR partially rescues the self-renewal defect caused by loss of Wnt signaling. Collectively, our findings support a model in which the Wnt and EGFR pathways operate in a signaling hierarchy to promote FSC self-renewal.


Subject(s)
Cell Self Renewal , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , ErbB Receptors/metabolism , Ovarian Follicle/cytology , Receptors, Invertebrate Peptide/metabolism , Stem Cells/cytology , Wnt1 Protein/metabolism , Animals , Drosophila Proteins/genetics , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphorylation , Receptors, Notch/metabolism , Signal Transduction , Stem Cell Niche , Stem Cells/metabolism , Transcription, Genetic
14.
EMBO Rep ; 18(12): 2105-2118, 2017 12.
Article in English | MEDLINE | ID: mdl-29158350

ABSTRACT

Understanding how cell fate decisions are regulated is a fundamental goal of developmental and stem cell biology. Most studies on the control of cell fate decisions address the contributions of changes in transcriptional programming, epigenetic modifications, and biochemical differentiation cues. However, recent studies have found that other aspects of cell biology also make important contributions to regulating cell fate decisions. These cues can have a permissive or instructive role and are integrated into the larger network of signaling, functioning both upstream and downstream of developmental signaling pathways. Here, we summarize recent insights into how cell fate decisions are influenced by four aspects of cell biology: metabolism, reactive oxygen species (ROS), intracellular pH (pHi), and cell morphology. For each topic, we discuss how these cell biological cues interact with each other and with protein-based mechanisms for changing gene transcription. In addition, we highlight several questions that remain unanswered in these exciting and relatively new areas of the field.


Subject(s)
Cell Differentiation/genetics , Signal Transduction/genetics , Stem Cells/metabolism , Cell Biology , Hydrogen-Ion Concentration , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Transcription, Genetic
15.
J Vis Exp ; (127)2017 09 26.
Article in English | MEDLINE | ID: mdl-28994781

ABSTRACT

Changes in intracellular pH (pHi) play important roles in the regulation of many cellular functions, including metabolism, proliferation, and differentiation. Typically, pHi dynamics are determined in cultured cells, which are amenable to measuring and experimentally manipulating pHi. However, the recent development of new tools and methodologies has made it possible to study pHi dynamics within intact, live tissue. For Drosophila research, one important development was the generation of a transgenic line carrying a pHi biosensor, mCherry::pHluorin. Here, we describe a protocol that we routinely use for imaging live Drosophila ovarioles to measure pHi in the epithelial follicle stem cell (FSC) lineage in mCherry::pHluorin transgenic wild type lines; however, the methods described here can be easily adapted for other tissues, including the wing discs and eye epithelium. We describe techniques for expressing mCherry::pHluorin in the FSC lineage, maintaining ovarian tissue during live imaging, and acquiring and analyzing images to obtain pHi values.


Subject(s)
Drosophila/physiology , Green Fluorescent Proteins/metabolism , Hydrogen-Ion Concentration , Ovarian Follicle/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Female , Ovarian Follicle/cytology , Stem Cells/cytology
16.
Genetics ; 206(3): 1417-1428, 2017 07.
Article in English | MEDLINE | ID: mdl-28512187

ABSTRACT

The process of selecting for cellular fitness through competition plays a critical role in both development and disease. The germarium, a structure at the tip of the ovariole of a Drosophila ovary, contains two follicle stem cells (FSCs) that undergo neutral competition for the stem cell niche. Using the FSCs as a model, we performed a genetic screen through a collection of 126 mutants in essential genes on the X chromosome to identify candidates that increase or decrease competition for the FSC niche. We identified ∼55 and 6% of the mutations screened as putative FSC hypo- or hyper-competitors, respectively. We found that a large majority of mutations in vesicle trafficking genes (11 out of the 13 in the collection of mutants) are candidate hypo-competition alleles, and we confirmed the hypo-competition phenotype for four of these alleles. We also show that Sec16 and another COPII vesicle trafficking component, Sar1, are required for follicle cell differentiation. Lastly, we demonstrate that, although some components of vesicle trafficking are also required for neutral competition in the cyst stem cells of the testis, there are important tissue-specific differences. Our results demonstrate a critical role for vesicle trafficking in stem cell niche competition and differentiation, and we identify a number of putative candidates for further exploration.


Subject(s)
Drosophila Proteins/metabolism , Ovarian Follicle/cytology , Stem Cell Niche , Testis/cytology , Vesicular Transport Proteins/metabolism , Animals , Cell Differentiation , Chromosomes, Insect/genetics , Drosophila/cytology , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Female , Male , Ovarian Follicle/metabolism , Testis/metabolism , Vesicular Transport Proteins/genetics , X Chromosome/genetics
17.
J Cell Biol ; 215(3): 345-355, 2016 Nov 07.
Article in English | MEDLINE | ID: mdl-27821494

ABSTRACT

Despite extensive knowledge about the transcriptional regulation of stem cell differentiation, less is known about the role of dynamic cytosolic cues. We report that an increase in intracellular pH (pHi) is necessary for the efficient differentiation of Drosophila adult follicle stem cells (FSCs) and mouse embryonic stem cells (mESCs). We show that pHi increases with differentiation from FSCs to prefollicle cells (pFCs) and follicle cells. Loss of the Drosophila Na+-H+ exchanger DNhe2 lowers pHi in differentiating cells, impairs pFC differentiation, disrupts germarium morphology, and decreases fecundity. In contrast, increasing pHi promotes excess pFC cell differentiation toward a polar/stalk cell fate through suppressing Hedgehog pathway activity. Increased pHi also occurs with mESC differentiation and, when prevented, attenuates spontaneous differentiation of naive cells, as determined by expression of microRNA clusters and stage-specific markers. Our findings reveal a previously unrecognized role of pHi dynamics for the differentiation of two distinct types of stem cell lineages, which opens new directions for understanding conserved regulatory mechanisms.


Subject(s)
Aging/physiology , Cell Differentiation , Drosophila melanogaster/cytology , Epithelial Cells/cytology , Intracellular Space/metabolism , Mouse Embryonic Stem Cells/cytology , Animals , Cell Lineage , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epithelial Cells/metabolism , Female , Hedgehog Proteins/metabolism , Hydrogen-Ion Concentration , Mice , Mouse Embryonic Stem Cells/metabolism , Ovarian Follicle/cytology , Signal Transduction
18.
Development ; 143(24): 4631-4642, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27836963

ABSTRACT

In the epithelial follicle stem cells (FSCs) of the Drosophila ovary, Epidermal Growth Factor Receptor (EGFR) signaling promotes self-renewal, whereas Notch signaling promotes differentiation of the prefollicle cell (pFC) daughters. We have identified two proteins, Six4 and Groucho (Gro), that link the activity of these two pathways to regulate the earliest cell fate decision in the FSC lineage. Our data indicate that Six4 and Gro promote differentiation towards the polar cell fate by promoting Notch pathway activity. This activity of Gro is antagonized by EGFR signaling, which inhibits Gro-dependent repression via p-ERK mediated phosphorylation. We have found that the phosphorylated form of Gro persists in newly formed pFCs, which may delay differentiation and provide these cells with a temporary memory of the EGFR signal. Collectively, these findings demonstrate that phosphorylated Gro labels a transition state in the FSC lineage and describe the interplay between Notch and EGFR signaling that governs the differentiation processes during this period.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , ErbB Receptors/metabolism , Homeodomain Proteins/genetics , Nerve Tissue Proteins/genetics , Ovarian Follicle/embryology , Receptors, Invertebrate Peptide/metabolism , Receptors, Notch/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Cell Differentiation/genetics , Epithelial Cells/cytology , Female , Homeodomain Proteins/metabolism , Nerve Tissue Proteins/metabolism , Ovarian Follicle/cytology , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/genetics , Stem Cells/cytology , Transcription Factors/metabolism
19.
Development ; 142(1): 82-91, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25516970

ABSTRACT

In the germarium of the Drosophila ovary, germline cysts are encapsulated one at a time by a follicular epithelium derived from two follicle stem cells (FSCs). Ovaries in flies mutant for the serine/threonine kinase Pak exhibit a novel phenotype, in which two side-by-side cysts are encapsulated at a time, generating paired egg chambers. This striking phenotype originates in the pupal ovary, where the developing germarium is shaped by the basal stalk, a stack of cells formed by cell intercalation. The process of basal stalk formation is not well understood, and we provide evidence that the cell intercalation is driven by actomyosin contractility of DE-Cadherin-adhered cells, leading to a column of disk-shaped cells exhibiting a novel radial cell polarity. Cell intercalation fails in Pak mutant ovaries, leading to abnormally wide basal stalks and consequently wide germaria with side-by-side cysts. We present evidence that Pak mutant germaria have extra FSCs, and we propose that contact of a germline cyst with the basal stalk in the pupal ovary contributes to FSC niche formation. The wide basal stalk in Pak mutants enables the formation of extra FSC niches which are mispositioned and yet functional, indicating that the FSC niche can be established in diverse locations.


Subject(s)
Cell Polarity , Drosophila melanogaster/cytology , Drosophila melanogaster/enzymology , Ovarian Follicle/cytology , Ovarian Follicle/enzymology , Stem Cell Niche , Animals , Drosophila Proteins , Drosophila melanogaster/anatomy & histology , Female , Models, Biological , Mutation/genetics , Ovum/cytology , Ovum/metabolism , Phenotype , p21-Activated Kinases
20.
Elife ; 32014 Dec 01.
Article in English | MEDLINE | ID: mdl-25437306

ABSTRACT

Epithelial stem cells divide asymmetrically, such that one daughter replenishes the stem cell pool and the other differentiates. We found that, in the epithelial follicle stem cell (FSC) lineage of the Drosophila ovary, epidermal growth factor receptor (EGFR) signaling functions specifically in the FSCs to promote the unique partially polarized state of the FSC, establish apical-basal polarity throughout the lineage, and promote FSC maintenance in the niche. In addition, we identified a novel connection between EGFR signaling and the cell-polarity regulator liver kinase B1 (LKB1), which indicates that EGFR signals through both the Ras-Raf-MEK-Erk pathway and through the LKB1-AMPK pathway to suppress apical identity. The development of apical-basal polarity is the earliest visible difference between FSCs and their daughters, and our findings demonstrate that the EGFR-mediated regulation of apical-basal polarity is essential for the segregation of stem cell and daughter cell fates.


Subject(s)
Cell Polarity , Drosophila/metabolism , ErbB Receptors/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Down-Regulation , Drosophila/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...