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 ; 151(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38063853

ABSTRACT

High-sugar diets (HSDs) often lead to obesity and type 2 diabetes, both metabolic syndromes associated with stem cell dysfunction. However, it is unclear whether excess dietary sugar affects stem cells. Here, we report that HSD impairs stem cell function in the intestine and ovaries of female Drosophila prior to the onset of insulin resistance, a hallmark of type 2 diabetes. Although 1 week of HSD leads to obesity, impaired oogenesis and altered lipid metabolism, insulin resistance does not occur. HSD increases glucose uptake by germline stem cells (GSCs) and triggers reactive oxygen species-induced JNK signaling, which reduces GSC proliferation. Removal of excess sugar from the diet reverses these HSD-induced phenomena. A similar phenomenon is found in intestinal stem cells (ISCs), except that HSD disrupts ISC maintenance and differentiation. Interestingly, tumor-like GSCs and ISCs are less responsive to HSD, which may be because of their dependence on glycolytic metabolism and high energy demand, respectively. This study suggests that excess dietary sugar induces oxidative stress and damages stem cells before insulin resistance develops, a mechanism that may also occur in higher organisms.


Subject(s)
Adult Stem Cells , Diabetes Mellitus, Type 2 , Drosophila Proteins , Insulin Resistance , Animals , Female , Drosophila/metabolism , Drosophila Proteins/metabolism , Reactive Oxygen Species/metabolism , Dietary Sugars/metabolism , Adult Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Obesity
2.
Cell Death Discov ; 9(1): 4, 2023 Jan 09.
Article in English | MEDLINE | ID: mdl-36617578

ABSTRACT

Tubulin s-palmitoylation involves the thioesterification of a cysteine residue in tubulin with palmitate. The palmitate moiety is produced by the fatty acid synthesis pathway, which is rate-limited by acetyl-CoA carboxylase (ACC). While it is known that ACC is phosphorylated at serine 79 (pSer79) by AMPK and accumulates at the spindle pole (SP) during mitosis, a functional role for tubulin palmitoylation during mitosis has not been identified. In this study, we found that modulating pSer79-ACC level at the SP using AMPK agonist and inhibitor induced spindle defects. Loss of ACC function induced spindle abnormalities in cell lines and in germ cells of the Drosophila germarium, and palmitic acid (PA) rescued the spindle defects in the cell line treated transiently with the ACC inhibitor, TOFA. Furthermore, inhibition of protein palmitoylating or depalmitoylating enzymes also induced spindle defects. Together, these data suggested that precisely regulated cellular palmitate level and protein palmitoylation may be required for accurate spindle assembly. We then showed that tubulin was largely palmitoylated in interphase cells but less palmitoylated in mitotic cells. TOFA treatment diminished tubulin palmitoylation at doses that disrupt microtubule (MT) instability and cause spindle defects. Moreover, spindle MTs comprised of α-tubulins mutated at the reported palmitoylation site exhibited disrupted dynamic instability. We also found that TOFA enhanced the MT-targeting drug-induced spindle abnormalities and cytotoxicity. Thus, our study reveals that precise regulation of ACC during mitosis impacts tubulin palmitoylation to delicately control MT dynamic instability and spindle assembly, thereby safeguarding nuclear and cell division.

3.
FEBS Open Bio ; 12(12): 2102-2110, 2022 12.
Article in English | MEDLINE | ID: mdl-36331359

ABSTRACT

Recent studies have shown that mitochondrial morphology can modulate organelle function and greatly affect stem cell behavior, thus affecting tissue homeostasis. As such, we previously showed that the accumulation of fragmented mitochondria in aged Drosophila ovarian germline stem cells (GSCs) contributes to age-dependent GSC loss. However, standard immunofluorescence methods to examine mitochondrial morphology yield images with insufficient resolution for rigorous analysis, while 3-dimensional electron microscopy examination of mitochondrial morphology is labor intensive and allows only limited sampling of mitochondria. To overcome these issues, we utilized the expansion microscopy technique to expand GSC samples by 4-fold in combination with mitochondrial immunofluorescence labeling. Here, we present a simple, inexpensive method for nanoscale optical imaging of mitochondria in the germline. This protocol may be beneficial for studies that require visualization of mitochondria or other fine subcellular structures in the Drosophila ovary.


Subject(s)
Drosophila Proteins , Oogonial Stem Cells , Animals , Female , Drosophila , Microscopy , Mitochondria
4.
Front Cell Dev Biol ; 10: 877047, 2022.
Article in English | MEDLINE | ID: mdl-35517512

ABSTRACT

Morphogen-mediated signaling is critical for proper organ development and stem cell function, and well-characterized mechanisms spatiotemporally limit the expression of ligands, receptors, and ligand-binding cell-surface glypicans. Here, we show that in the developing Drosophila ovary, canonical Wnt signaling promotes the formation of somatic escort cells (ECs) and their protrusions, which establish a physical permeability barrier to define morphogen territories for proper germ cell differentiation. The protrusions shield germ cells from Dpp and Wingless morphogens produced by the germline stem cell (GSC) niche and normally only received by GSCs. Genetic disruption of EC protrusions allows GSC progeny to also receive Dpp and Wingless, which subsequently disrupt germ cell differentiation. Our results reveal a role for canonical Wnt signaling in specifying the ovarian somatic cells necessary for germ cell differentiation. Additionally, we demonstrate the morphogen-limiting function of this physical permeability barrier, which may be a common mechanism in other organs across species.

5.
Development ; 148(15)2021 08 01.
Article in English | MEDLINE | ID: mdl-34323273

ABSTRACT

Vertebrate animals usually display robust growth trajectories during juvenile stages, and reversible suspension of this growth momentum by a single genetic determinant has not been reported. Here, we report a single genetic factor that is essential for juvenile growth in zebrafish. Using a forward genetic screen, we recovered a temperature-sensitive allele, pan (after Peter Pan), that suspends whole-organism growth at juvenile stages. Remarkably, even after growth is halted for a full 8-week period, pan mutants are able to resume a robust growth trajectory after release from the restrictive temperature, eventually growing into fertile adults without apparent adverse phenotypes. Positional cloning and complementation assays revealed that pan encodes a probable ATP-dependent RNA helicase (DEAD-Box Helicase 52; ddx52) that maintains the level of 47S precursor ribosomal RNA. Furthermore, genetic silencing of ddx52 and pharmacological inhibition of bulk RNA transcription similarly suspend the growth of flies, zebrafish and mice. Our findings reveal evidence that safe, reversible pauses of juvenile growth can be mediated by targeting the activity of a single gene, and that its pausing mechanism has high evolutionary conservation.


Subject(s)
RNA Helicases/genetics , RNA/genetics , Zebrafish/genetics , Alleles , Animals , Female , Gene Silencing/physiology , Male , Mice , Mice, Inbred C57BL , RNA Precursors/genetics , Ribosomes/genetics , Transcription, Genetic/genetics
6.
Aging Cell ; 19(8): e13191, 2020 08.
Article in English | MEDLINE | ID: mdl-32666649

ABSTRACT

Changes in mitochondrial dynamics (fusion and fission) are known to occur during stem cell differentiation; however, the role of this phenomenon in tissue aging remains unclear. Here, we report that mitochondrial dynamics are shifted toward fission during aging of Drosophila ovarian germline stem cells (GSCs), and this shift contributes to aging-related GSC loss. We found that as GSCs age, mitochondrial fragmentation and expression of the mitochondrial fission regulator, Dynamin-related protein (Drp1), are both increased, while mitochondrial membrane potential is reduced. Moreover, preventing mitochondrial fusion in GSCs results in highly fragmented depolarized mitochondria, decreased BMP stemness signaling, impaired fatty acid metabolism, and GSC loss. Conversely, forcing mitochondrial elongation promotes GSC attachment to the niche. Importantly, maintenance of aging GSCs can be enhanced by suppressing Drp1 expression to prevent mitochondrial fission or treating with rapamycin, which is known to promote autophagy via TOR inhibition. Overall, our results show that mitochondrial dynamics are altered during physiological aging, affecting stem cell homeostasis via coordinated changes in stemness signaling, niche contact, and cellular metabolism. Such effects may also be highly relevant to other stem cell types and aging-induced tissue degeneration.


Subject(s)
Adult Germline Stem Cells/metabolism , Mitochondrial Dynamics/genetics , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Drosophila , Female , Male , Signal Transduction
7.
Nat Commun ; 11(1): 3147, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561720

ABSTRACT

Transposons are known to participate in tissue aging, but their effects on aged stem cells remain unclear. Here, we report that in the Drosophila ovarian germline stem cell (GSC) niche, aging-related reductions in expression of Piwi (a transposon silencer) derepress retrotransposons and cause GSC loss. Suppression of Piwi expression in the young niche mimics the aged niche, causing retrotransposon depression and coincident activation of Toll-mediated signaling, which promotes Glycogen synthase kinase 3 activity to degrade ß-catenin. Disruption of ß-catenin-E-cadherin-mediated GSC anchorage then results in GSC loss. Knocking down gypsy (a highly active retrotransposon) or toll, or inhibiting reverse transcription in the piwi-deficient niche, suppresses GSK3 activity and ß-catenin degradation, restoring GSC-niche attachment. This retrotransposon-mediated impairment of aged stem cell maintenance may have relevance in many tissues, and could represent a viable therapeutic target for aging-related tissue degeneration.


Subject(s)
Argonaute Proteins/metabolism , Cellular Senescence , Drosophila Proteins/metabolism , Drosophila melanogaster , Germ Cells/metabolism , Animals , Argonaute Proteins/genetics , Cadherins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Gene Silencing , Glycogen Synthase Kinase 3/metabolism , Ovary/cytology , Ovary/metabolism , Retroelements/genetics , Signal Transduction , Stem Cell Niche/physiology , Stem Cells/metabolism , Toll-Like Receptors/metabolism , beta Catenin/metabolism
8.
Curr Opin Insect Sci ; 37: 16-22, 2020 02.
Article in English | MEDLINE | ID: mdl-32070932

ABSTRACT

Insect oogenesis is greatly affected by nutrient availability. When nutrients are abundant, oocytes are rapidly generated, but the process is slowed to conserve energy under nutrient-deficient conditions. To properly allocate limited resources toward oogenesis, systemic factors coordinate the behavioral response of ovarian germline stem cells (GSCs) to nutritional inputs by acting on the GSC itself, GSC supporting cells (the niche), or the adipose tissue surrounding the ovary. In this review, we describe current knowledge of the Drosophila ovarian GSC-niche-adipocyte system and major nutrient sensing pathways (insulin/IGF signaling, TOR signaling, and GCN2-dependent amino acid sensing) that intrinsically or extrinsically regulate GSC responses to nutrient signals.


Subject(s)
Drosophila/physiology , Oogonial Stem Cells/metabolism , Signal Transduction , Adipocytes/metabolism , Animals , Female , Oogonial Stem Cells/physiology , Stem Cell Niche/physiology
9.
Development ; 147(2)2020 01 15.
Article in English | MEDLINE | ID: mdl-31941704

ABSTRACT

WD40 proteins control many cellular processes via protein interactions. Drosophila Wuho (Wh, a WD40 protein) controls fertility, although the involved mechanisms are unclear. Here, we show that Wh promotion of Mei-p26 (a human TRIM32 ortholog) function maintains ovarian germ cell homeostasis. Wh and Mei-p26 are epistatically linked, with wh and mei-p26 mutants showing nearly identical phenotypes, including germline stem cell (GSC) loss, stem-cyst formation due to incomplete cytokinesis between GSCs and daughter cells, and overproliferation of GSC progeny. Mechanistically, Wh interacts with Mei-p26 in different cellular contexts to induce cell type-specific effects. In GSCs, Wh and Mei-p26 promote BMP stemness signaling for proper GSC division and maintenance. In GSC progeny, Wh and Mei-p26 silence nanos translation, downregulate a subset of microRNAs involved in germ cell differentiation and suppress ribosomal biogenesis via dMyc to limit germ cell mitosis. We also found that the human ortholog of Wh (WDR4) interacts with TRIM32 in human cells. Our results show that Wh is a regulator of Mei-p26 in Drosophila germ cells and suggest that the WD40-TRIM interaction may also control tissue homeostasis in other stem cell systems.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Germ Cells/metabolism , Homeostasis , Animals , Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Conserved Sequence , Drosophila melanogaster/cytology , Evolution, Molecular , Female , Fertility , Germ Cells/cytology , Meiosis , MicroRNAs/genetics , MicroRNAs/metabolism , Mitosis , Models, Biological , Mutation/genetics , Ovary/cytology , Ovum/cytology , Ovum/metabolism , Phenotype , Protein Binding , Ribosomes/metabolism , Signal Transduction
10.
Cell Mol Life Sci ; 76(21): 4309-4317, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31300869

ABSTRACT

Adult stem cells have a unique capacity to renew themselves and generate differentiated cells that are needed in the body. These cells are recruited and maintained by the surrounding microenvironment, known as the stem cell niche, during organ development. Thus, the stem cell niche is required for proper tissue homeostasis, and its dysregulation is associated with tumorigenesis and tissue degeneration. The identification of niche components and the mechanisms that regulate niche establishment and maintenance, however, are just beginning to be uncovered. Germline stem cells (GSCs) of the Drosophila ovary provide an excellent model for studying the stem cell niche in vivo because of their well-characterized cell biology and the availability of genetic tools. In this review, we introduce the ovarian GSC niche, and the key signaling pathways for niche precursor segregation, niche specification, and niche extracellular environment establishment and niche maintenance that are involved in regulating niche size during development and adulthood.


Subject(s)
Drosophila melanogaster , Oogonial Stem Cells/cytology , Stem Cell Niche/genetics , Animals , Cell Differentiation/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/physiology , Female , Germ Cells/cytology , Germ Cells/physiology , Oogonial Stem Cells/physiology , Ovary/cytology , Signal Transduction/genetics
11.
G3 (Bethesda) ; 9(6): 2007-2016, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31018943

ABSTRACT

The stem cell niche, a regulatory microenvironment, houses and regulates stem cells for maintenance of tissues throughout an organism's lifespan. While it is known that stem cell function declines with age, the role of niche cells in this decline is not completely understood. Drosophila exhibits a short lifespan with well-characterized ovarian germline stem cells (GSCs) and niche compartments, providing a good model with which to study stem cell biology. However, no inducible tools for temporal and spatial control of gene expression in the GSC-niche unit have been previously developed for aging studies. The current UAS-GAL4 systems are not ideal for aging studies because fly physiological aging may be affected by the temperature shifts used to manipulate GAL4 activity. Additionally, the actual needs of the aged niche may be masked by continuously driven gene expression. Since GeneSwitch GAL4 is conveniently activated by the steroid RU486 (mifepristone), we conducted an enhancer-trap screen to isolate GeneSwitch GAL4 lines with expression in the GSC-niche unit. We identified six lines with expression in germarial somatic cells, and two lines (#2305 and #2261) with expression in niche cap cells, the major constituent of the GSC niche. The use of lines #2305 or #2261 to overexpress Drosophila insulin-like peptide 2, which maintains GSC lifespan, in aged niche cap cells significantly delayed age-dependent GSC loss. These results support the notion that insulin signaling is beneficial for maintaining aged stem cells and also validate the utility of our GeneSwitch GAL4 lines for studying stem cell aging.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Gene Expression , Oogonial Stem Cells/metabolism , Stem Cell Niche/genetics , Transcription Factors/genetics , Animals , Animals, Genetically Modified , Crosses, Genetic , Drosophila Proteins/metabolism , Female , Fluorescent Antibody Technique , Gene Order , Genetic Vectors/genetics , Immunohistochemistry , Male , Phenotype , Quantitative Trait Loci , Transcription Factors/metabolism
12.
Stem Cell Reports ; 11(3): 811-827, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30122445

ABSTRACT

In developing organisms, proper tuning of the number of stem cells within a niche is critical for the maintenance of adult tissues; however, the involved mechanisms remain largely unclear. Here, we demonstrate that Thickveins (Tkv), a type I bone morphogenetic protein (BMP) receptor, acts in the Drosophila developing ovarian soma through a Smad-independent pathway to shape the distribution of BMP signal within the niche, impacting germline stem cell (GSC) recruitment and maintenance. Somatic Tkv promotes Egfr signaling to silence transcription of Dally, which localizes BMP signals on the cell surface. In parallel, Tkv promotes Hh signaling, which promotes escort cell cellular protrusions and upregulates expression of the Drosophila BMP homolog, Dpp, forming a positive feedback loop that enhances Tkv signaling and strengthens the niche boundary. Our results reveal a role for non-canonical BMP signaling in the soma during GSC establishment and generally illustrate how complex, cell-specific BMP signaling mediates niche-stem cell interactions.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Germ Cells/cytology , Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Smad Proteins/metabolism , Animals , Cell Differentiation , Drosophila/cytology , Drosophila/growth & development , Female , Germ Cells/metabolism , Male , Ovary/cytology , Ovary/growth & development , Ovary/metabolism , Stem Cell Niche
13.
G3 (Bethesda) ; 8(7): 2345-2354, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29764959

ABSTRACT

Adult stem cells maintain tissue homeostasis. This unique capability largely depends on the stem cell niche, a specialized microenvironment, which preserves stem cell identity through physical contacts and secreted factors. In many cancers, latent tumor cell niches are thought to house stem cells and aid tumor initiation. However, in developing tissue and cancer it is unclear how the niche is established. The well-characterized germline stem cells (GSCs) and niches in the Drosophila melanogaster ovary provide an excellent model to address this fundamental issue. As such, we conducted a small-scale RNAi screen of 560 individually expressed UAS-RNAi lines with targets implicated in female fertility. RNAi was expressed in the soma of larval gonads, and screening for reduced egg production and abnormal ovarian morphology was performed in adults. Twenty candidates that affect ovarian development were identified and subsequently knocked down in the soma only during niche formation. Feminization factors (Transformer, Sex lethal, and Virilizer), a histone methyltransferase (Enhancer of Zeste), a transcriptional machinery component (Enhancer of yellow 1), a chromatin remodeling complex member (Enhancer of yellow 3) and a chromosome passenger complex constituent (Incenp) were identified as potentially functioning in the control of niche size. The identification of these molecules highlights specific molecular events that are critical for niche formation and will provide a basis for future studies to fully understand the mechanisms of GSC recruitment and maintenance.


Subject(s)
Drosophila/genetics , Gene Expression Regulation , Germ Cells/metabolism , Infertility, Female/genetics , Stem Cell Niche , Animals , Cell Differentiation , Female , Gene Knockdown Techniques , Genetic Testing/methods , Male , Ovary/cytology , Ovary/metabolism , Phenotype , RNA Interference , RNA, Small Interfering/genetics
14.
Development ; 145(7)2018 04 05.
Article in English | MEDLINE | ID: mdl-29549109

ABSTRACT

Diet is an important regulator of stem cell homeostasis; however, the underlying mechanisms of this regulation are not fully known. Here, we report that insulin signaling mediates dietary maintenance of Drosophila ovarian germline stem cells (GSCs) by promoting the extension of niche escort cell (EC) membranes to wrap around GSCs. This wrapping may facilitate the delivery of bone morphogenetic protein stemness factors from ECs in the niche to GSCs. In addition to the effects on GSCs, insulin signaling-mediated regulation of EC number and protrusions controls the division and growth of GSC progeny. The effects of insulin signaling on EC membrane extension are, at least in part, driven by enhanced translation of Failed axon connections (Fax) via Ribosomal protein S6 kinase. Fax is a membrane protein that may participate in Abelson tyrosine kinase-regulated cytoskeletal dynamics and is known to be involved in axon bundle formation. Therefore, we conclude that dietary cues stimulate insulin signaling in the niche to regulate EC cellular structure, probably via Fax-dependent cytoskeleton remodeling. This mechanism enhances intercellular contact and facilitates homeostatic interactions between somatic and germline cells in response to diet.


Subject(s)
Cell Surface Extensions/physiology , Diet , Germ Cells/physiology , Homeostasis/physiology , Insulin/metabolism , Stem Cell Niche/physiology , Animals , Blotting, Western , Cell Survival/physiology , Cues , Drosophila/cytology , Drosophila/metabolism , Drosophila/physiology , Drosophila Proteins/metabolism , Female , Fluorescent Antibody Technique , Germ Cells/cytology , Germ Cells/metabolism , Ovary/metabolism , Ovary/physiology , Real-Time Polymerase Chain Reaction , Signal Transduction
15.
PLoS One ; 12(12): e0188917, 2017.
Article in English | MEDLINE | ID: mdl-29261681

ABSTRACT

Snail, a zinc-finger transcription factor, controls the process of epithelial-mesenchymal transition, and ectopic expression of this protein may produce cells with stem cell properties. Because the effect of Snail expression in ovarian epithelial cells remains unclear, we generated Drosophila ovarian follicle stem cells (FSCs) with homozygous Scutoid (Sco) mutation. The Sco mutation is a reciprocal transposition that is known to induce ectopic Snail activity. We found that Sco mutant FSCs showed excess proliferation and high competitiveness for niche occupancy, and the descendants of this lineage formed outgrowths that failed to enter the endocycle. Surprisingly, such phenotypes were not rescued by suppressing Snail expression, but were completely restored by supplying Lethal giant larvae (Lgl). The lgl allele is a cell polarity gene that is often mutated in the genome. Importantly, Sco mutants survived in a complementation test with lgl. This result was probably obtained because the Sco-associated lgl allele appears to diminish, but not ablate lgl expression. While our data do not rule out the possibility that the Sco mutation disrupts a regulator of lgl transcription, our results strongly suggest that the phenotypes we found in Sco mutants are more closely associated with the lgl allele than ectopic Snail activity.


Subject(s)
Drosophila Proteins/genetics , Drosophila/growth & development , Gene Expression Regulation , Ovarian Follicle/cytology , Tumor Suppressor Proteins/genetics , Alleles , Animals , Cell Lineage , Cell Proliferation/genetics , Cellular Senescence , Drosophila/genetics , Female , Homeostasis , Stem Cells/cytology
16.
J Cell Biol ; 216(5): 1439-1453, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28363970

ABSTRACT

Stem cells require different types of supporting cells, or niches, to control stem cell maintenance and differentiation. However, little is known about how those niches are formed. We report that in the development of the Drosophila melanogaster ovary, the Hedgehog (Hh) gradient sets differential cell affinity for somatic gonadal precursors to specify stromal intermingled cells, which contributes to both germline stem cell maintenance and differentiation niches in the adult. We also report that Traffic Jam (an orthologue of a large Maf transcription factor in mammals) is a novel transcriptional target of Hh signaling to control cell-cell adhesion by negative regulation of E-cadherin expression. Our results demonstrate the role of Hh signaling in niche establishment by segregating somatic cell lineages for differentiation.


Subject(s)
Cell Adhesion , Germ Cells/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Stem Cell Niche , Animals , Drosophila melanogaster , Female , Ovary/cytology , Ovary/metabolism
17.
Int J Mol Sci ; 18(1)2017 Jan 11.
Article in English | MEDLINE | ID: mdl-28085063

ABSTRACT

Paclobutrazol (PBZ) is a widely used fungicide that shows toxicity to aquatic embryos, probably through rain-wash. Here, we specifically focus on its toxic effect on eye development in zebrafish, as well as the role of retinoic acid (RA), a metabolite of vitamin A that controls proliferation and differentiation of retinal photoreceptor cells, in this toxicity. Embryos were exposed to PBZ with or without RA from 2 to 72 h post-fertilization (hpf), and PBZ-treated embryos (2-72 hpf) were exposed to RA for additional hours until 120 hpf. Eye size and histology were examined. Expression levels of gnat1 (rod photoreceptor marker), gnat2 (cone photoreceptor marker), aldehyde dehydrogenases (encoding key enzymes for RA synthesis), and phospho-histone H3 (an M-phase marker) in the eyes of control and treated embryos were examined. PBZ exposure dramatically reduces photoreceptor proliferation, thus resulting in a thinning of the photoreceptor cell layer and leading to a small eye. Co-treatment of PBZ with RA, or post-treatment of PBZ-treated embryos with RA, partially rescues photoreceptor cells, revealed by expression levels of marker proteins and by retinal cell proliferation. PBZ has strong embryonic toxicity to retinal photoreceptors, probably via suppressing the production of RA, with effects including impaired retinal cell division.


Subject(s)
Cell Differentiation/drug effects , Neuroprotective Agents/pharmacology , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/drug effects , Tretinoin/pharmacology , Triazoles/toxicity , Animals , Dose-Response Relationship, Drug , Embryo, Nonmammalian , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/pathology , Tretinoin/metabolism , Zebrafish
18.
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
19.
Dev Biol ; 414(2): 142-8, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27141871

ABSTRACT

Epithelial stem cells undergo constant self-renewal and differentiation to maintain the homeostasis of epithelial tissues that undergo rapid turnover. Recent studies have shown that the epithelial-mesenchymal transition (EMT), which is primarily mediated by Snail via the suppression of E-cadherin, is able to generate cells with stem cell properties. However, the role of Snail in epithelial stem cells remains unclear. Here, we report that Snail directly controls proliferation of follicle stem cells (FSCs) in Drosophila females. Disruption of Snail expression in FSCs compromises their proliferation, but not their maintenance. Conversely, FSCs with excessive Snail expression display increased proliferation and lifespan, which is accompanied by a moderate decrease in the expression of E-cadherin (required for adhesion of FSCs to their niche) at the junction between their adjacent cells, indicating a conserved role of Snail in E-cadherin inhibition, which promote epithelial cell proliferation. Interestingly, a decrease in E-cadherin in snail-knock down FSCs does not restore the decreased proliferation of snail-knock down FSCs, suggesting that adhesion strength of FSCs to their niche is dispensable for Snail-mediated FSC division. Our results demonstrate that Snail controls epithelial stem cell division independently of its known role in the EMT, which contributes to induction of cancer stem cells.


Subject(s)
Cadherins/biosynthesis , Drosophila Proteins/biosynthesis , Ovarian Follicle/cytology , Ovary/cytology , Snail Family Transcription Factors/physiology , Stem Cells/cytology , Animals , Cadherins/genetics , Cell Adhesion , Cell Division , Cellular Senescence , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Epithelial Cells/cytology , Epithelial-Mesenchymal Transition/physiology , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Luminescent Proteins/analysis , Mosaicism , Snail Family Transcription Factors/deficiency , Stem Cell Niche
SELECTION OF CITATIONS
SEARCH DETAIL
...