Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 153
Filter
1.
PLoS Biol ; 22(10): e3002823, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39401187

ABSTRACT

Epithelial tissues undergo cell turnover both during development and for homeostatic maintenance. Removal of cells is coordinated with the increase in number of newly dividing cells to maintain barrier function of the tissue. In Drosophila metamorphosis, larval epidermal cells (LECs) are replaced by adult precursor cells called histoblasts. Removal of LECs must counterbalance the exponentially increasing adult histoblasts. Previous work showed that the LEC removal accelerates as endocytic activity decreases throughout all LECs. Here, we show that the acceleration is accompanied by a mode switching from isolated single-cell apoptosis to clustered ones induced by the endocytic activity reduction. We identify the epidermal growth factor receptor (EGFR) pathway via extracellular-signal regulated kinase (ERK) activity as the main components downstream of endocytic activity in LECs. The reduced ERK activity, caused by the decrease in endocytic activity, is responsible for the apoptotic mode switching. Initially, ERK is transiently activated in normal LECs surrounding a single apoptotic LEC in a ligand-dependent manner, preventing clustered cell death. Following the reduction of endocytic activity, LEC apoptosis events do not provoke these transient ERK up-regulations, resulting in the acceleration of the cell elimination rate by frequent clustered apoptosis. These findings contrasted with the common perspective that clustered apoptosis is disadvantageous. Instead, switching to clustered apoptosis is required to accommodate the growth of neighboring tissues.


Subject(s)
Apoptosis , Drosophila Proteins , Drosophila melanogaster , Endocytosis , ErbB Receptors , Signal Transduction , Animals , Endocytosis/physiology , ErbB Receptors/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Larva/metabolism , Metamorphosis, Biological/physiology , Receptors, Invertebrate Peptide/metabolism , Receptors, Invertebrate Peptide/genetics , Epithelium/metabolism , Epidermal Cells/metabolism , Drosophila/metabolism
2.
PLoS Biol ; 22(7): e3002547, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39047051

ABSTRACT

Despite the deep conservation of the DNA damage response (DDR) pathway, cells in different contexts vary widely in their susceptibility to DNA damage and their propensity to undergo apoptosis as a result of genomic lesions. One of the cell signaling pathways implicated in modulating the DDR is the highly conserved Wnt pathway, which is known to promote resistance to DNA damage caused by ionizing radiation in a variety of human cancers. However, the mechanisms linking Wnt signal transduction to the DDR remain unclear. Here, we use a genetically encoded system in Drosophila to reliably induce consistent levels of DNA damage in vivo, and demonstrate that canonical Wnt signaling in the wing imaginal disc buffers cells against apoptosis in the face of DNA double-strand breaks. We show that Wg, the primary Wnt ligand in Drosophila, activates epidermal growth factor receptor (EGFR) signaling via the ligand-processing protease Rhomboid, which, in turn, modulates the DDR in a Chk2-, p53-, and E2F1-dependent manner. These studies provide mechanistic insight into the modulation of the DDR by the Wnt and EGFR pathways in vivo in a highly proliferative tissue. Furthermore, they reveal how the growth and patterning functions of Wnt signaling are coupled with prosurvival, antiapoptotic activities, thereby facilitating developmental robustness in the face of genomic damage.


Subject(s)
Apoptosis , DNA Damage , Drosophila Proteins , ErbB Receptors , Imaginal Discs , Wings, Animal , Wnt Signaling Pathway , Wnt1 Protein , Animals , ErbB Receptors/metabolism , ErbB Receptors/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Wings, Animal/metabolism , Wings, Animal/growth & development , Imaginal Discs/metabolism , Imaginal Discs/growth & development , Wnt1 Protein/metabolism , Wnt1 Protein/genetics , Apoptosis/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/growth & development , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Checkpoint Kinase 2/metabolism , Checkpoint Kinase 2/genetics , Signal Transduction , DNA Breaks, Double-Stranded , Receptors, Invertebrate Peptide/metabolism , Receptors, Invertebrate Peptide/genetics , Drosophila/metabolism , Drosophila/genetics , Transcription Factors
3.
Nat Commun ; 15(1): 5493, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38944657

ABSTRACT

JNK signaling is a critical regulator of inflammation and regeneration, but how it is controlled in specific tissue contexts remains unclear. Here we show that, in the Drosophila intestine, the TNF-type ligand, Eiger (Egr), is expressed exclusively by intestinal stem cells (ISCs) and enteroblasts (EBs), where it is induced by stress and during aging. Egr preferentially activates JNK signaling in a paracrine fashion in differentiated enterocytes (ECs) via its receptor, Grindelwald (Grnd). N-glycosylation genes (Alg3, Alg9) restrain this activation, and stress-induced downregulation of Alg3 and Alg9 correlates with JNK activation, suggesting a regulatory switch. JNK activity in ECs induces expression of the intermembrane protease Rhomboid (Rho), driving secretion of EGFR ligands Keren (Krn) and Spitz (Spi), which in turn activate EGFR signaling in progenitor cells (ISCs and EBs) to stimulate their growth and division, as well as to produce more Egr. This study uncovers an N-glycosylation-controlled, paracrine JNK-EGFR-JNK feedforward loop that sustains ISC proliferation during stress-induced gut regeneration.


Subject(s)
Drosophila Proteins , ErbB Receptors , Intestines , MAP Kinase Signaling System , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , ErbB Receptors/metabolism , ErbB Receptors/genetics , Intestines/cytology , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Enterocytes/metabolism , Enterocytes/cytology , Stem Cells/metabolism , Stem Cells/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Drosophila/metabolism , Glycosylation , Receptors, Invertebrate Peptide/metabolism , Receptors, Invertebrate Peptide/genetics , Cell Proliferation , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction , Cell Communication , Cell Differentiation , Epidermal Growth Factor , Membrane Proteins
4.
Genetics ; 227(1)2024 05 07.
Article in English | MEDLINE | ID: mdl-38551457

ABSTRACT

Across diverse insect taxa, the behavior and physiology of females dramatically changes after mating-processes largely triggered by the transfer of seminal proteins from their mates. In the vinegar fly Drosophila melanogaster, the seminal protein sex peptide (SP) decreases the likelihood of female flies remating and causes additional behavioral and physiological changes that promote fertility including increasing egg production. Although SP is only found in the Drosophila genus, its receptor, sex peptide receptor (SPR), is the widely conserved myoinhibitory peptide (MIP) receptor. To test the functional role of SPR in mediating postmating responses in a non-Drosophila dipteran, we generated 2 independent Spr-knockout alleles in the yellow fever mosquito, Aedes aegypti. Although SPR is needed for postmating responses in Drosophila and the cotton bollworm Helicoverpa armigera, Spr mutant Ae. aegypti show completely normal postmating decreases in remating propensity and increases in egg laying. In addition, injection of synthetic SP or accessory gland homogenate from D. melanogaster into virgin female mosquitoes did not elicit these postmating responses. Our results demonstrate that Spr is not required for these canonical postmating responses in Ae. aegypti, indicating that other, as yet unknown, signaling pathways are likely responsible for these behavioral switches in this disease vector.


Subject(s)
Aedes , Insect Proteins , Oviposition , Receptors, Invertebrate Peptide , Animals , Female , Male , Aedes/genetics , Aedes/physiology , Drosophila melanogaster/physiology , Drosophila melanogaster/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Insect Proteins/genetics , Insect Proteins/metabolism , Receptors, Invertebrate Peptide/metabolism , Receptors, Invertebrate Peptide/genetics , Sexual Behavior, Animal
5.
PLoS Genet ; 19(2): e1010622, 2023 02.
Article in English | MEDLINE | ID: mdl-36730442

ABSTRACT

The Epidermal Growth Factor Receptor (EGFR) signaling pathway plays a critical role in regulating tissue patterning. Drosophila EGFR signaling achieves specificity through multiple ligands and feedback loops to finetune signaling outcomes spatiotemporally. The principal Drosophila EGF ligand, cleaved Spitz, and the negative feedback regulator, Argos are diffusible and can act both in a cell autonomous and non-autonomous manner. The expression dose of Spitz and Argos early in photoreceptor cell fate determination has been shown to be critical in patterning the Drosophila eye, but the exact identity of the cells expressing these genes in the larval eye disc has been elusive. Using single molecule RNA Fluorescence in situ Hybridization (smFISH), we reveal an intriguing differential expression of spitz and argos mRNA in the Drosophila third instar eye imaginal disc indicative of directional non-autonomous EGFR signaling. By genetically tuning EGFR signaling, we show that rather than absolute levels of expression, the ratio of expression of spitz-to-argos to be a critical determinant of the final adult eye phenotype. Proximate effects on EGFR signaling in terms of cell cycle and differentiation markers are affected differently in the different perturbations. Proper ommatidial patterning is robust to thresholds around a tightly maintained wildtype spitz-to-argos ratio, and breaks down beyond. This provides a powerful instance of developmental buffering against gene expression fluctuations.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , In Situ Hybridization, Fluorescence , Epidermal Growth Factor/genetics , Signal Transduction/genetics , Eye/metabolism , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism
6.
Curr Biol ; 32(17): 3704-3719.e7, 2022 09 12.
Article in English | MEDLINE | ID: mdl-35896119

ABSTRACT

EGFR-RAS-ERK signaling promotes growth and proliferation in many cell types, and genetic hyperactivation of RAS-ERK signaling drives many cancers. Yet, despite intensive study of upstream components in EGFR signal transduction, the identities and functions of downstream effectors in the pathway are poorly understood. In Drosophila intestinal stem cells (ISCs), the transcriptional repressor Capicua (Cic) and its targets, the ETS-type transcriptional activators Pointed (pnt) and Ets21C, are essential downstream effectors of mitogenic EGFR signaling. Here, we show that these factors promote EGFR-dependent metabolic changes that increase ISC mass, mitochondrial growth, and mitochondrial activity. Gene target analysis using RNA and DamID sequencing revealed that Pnt and Ets21C directly upregulate not only DNA replication and cell cycle genes but also genes for oxidative phosphorylation, the TCA cycle, and fatty acid beta-oxidation. Metabolite analysis substantiated these metabolic functions. The mitochondrial transcription factor B2 (mtTFB2), a direct target of Pnt, was required and partially sufficient for EGFR-driven ISC growth, mitochondrial biogenesis, and proliferation. MEK-dependent EGF signaling stimulated mitochondrial biogenesis in human RPE-1 cells, indicating the conservation of these metabolic effects. This work illustrates how EGFR signaling alters metabolism to coordinately activate cell growth and cell division.


Subject(s)
Drosophila Proteins , Animals , Cell Proliferation , DNA-Binding Proteins/metabolism , Drosophila/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Nerve Tissue Proteins , Organelle Biogenesis , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-ets/metabolism , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Transcription Factors/metabolism
7.
PLoS Genet ; 18(6): e1010224, 2022 06.
Article in English | MEDLINE | ID: mdl-35666718

ABSTRACT

How cell to cell interactions control local tissue growth to attain a species-specific organ size is a central question in developmental biology. The Drosophila Neural Cell Adhesion Molecule, Fasciclin 2, is expressed during the development of neural and epithelial organs. Fasciclin 2 is a homophilic-interaction protein that shows moderate levels of expression in the proliferating epithelia and high levels in the differentiating non-proliferative cells of imaginal discs. Genetic interactions and mosaic analyses reveal a cell autonomous requirement of Fasciclin 2 to promote cell proliferation in imaginal discs. This function is mediated by the EGFR, and indirectly involves the JNK and Hippo signaling pathways. We further show that Fasciclin 2 physically interacts with EGFR and that, in turn, EGFR activity promotes the cell autonomous expression of Fasciclin 2 during imaginal disc growth. We propose that this auto-stimulatory loop between EGFR and Fasciclin 2 is at the core of a cell to cell interaction mechanism that controls the amount of intercalary growth in imaginal discs.


Subject(s)
Drosophila Proteins , Imaginal Discs , Animals , Cell Proliferation/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , ErbB Receptors/genetics , Receptors, Invertebrate Peptide/genetics , Wings, Animal
8.
Elife ; 112022 04 25.
Article in English | MEDLINE | ID: mdl-35468055

ABSTRACT

Adult stem cells are maintained in niches, specialized microenvironments that regulate their self-renewal and differentiation. In the adult Drosophila testis stem cell niche, somatic hub cells produce signals that regulate adjacent germline stem cells (GSCs) and somatic cyst stem cells (CySCs). Hub cells are normally quiescent, but after complete genetic ablation of CySCs, they can proliferate and transdifferentiate into new CySCs. Here we find that Epidermal growth factor receptor (EGFR) signaling is upregulated in hub cells after CySC ablation and that the ability of testes to recover from ablation is inhibited by reduced EGFR signaling. In addition, activation of the EGFR pathway in hub cells is sufficient to induce their proliferation and transdifferentiation into CySCs. We propose that EGFR signaling, which is normally required in adult cyst cells, is actively inhibited in adult hub cells to maintain their fate but is repurposed to drive stem cell regeneration after CySC ablation.


Subject(s)
Cysts , Drosophila Proteins , Animals , Cell Transdifferentiation , Cysts/metabolism , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Male , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Stem Cells/physiology , Testis/metabolism , Tumor Microenvironment
9.
Stem Cell Reports ; 17(5): 1120-1137, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35427486

ABSTRACT

Adult tissue homeostasis is maintained by residential stem cells. The proliferation and differentiation of adult stem cells must be tightly balanced to avoid excessive proliferation or premature differentiation. However, how stem cell proliferation is properly controlled remains elusive. Here, we find that auxilin (Aux) restricts intestinal stem cell (ISC) proliferation mainly through EGFR signaling. aux depletion leads to excessive ISC proliferation and midgut homeostasis disruption, which is unlikely caused by defective Notch signaling. Aux is expressed in multiple types of intestinal cells. Interestingly, aux depletion causes a dramatic increase in EGFR signaling, with a strong accumulation of EGFR at the plasma membrane and an increased expression of EGFR ligands in response to tissue stress. Furthermore, Aux co-localizes and associates with EGFR. Finally, blocking EGFR signaling completely suppresses the defects caused by aux depletion. Together, these data demonstrate that Aux mainly safeguards EGFR activation to keep a proper ISC proliferation rate to maintain midgut homeostasis.


Subject(s)
Drosophila Proteins , Animals , Auxilins/metabolism , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila melanogaster , ErbB Receptors/metabolism , Intestines , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism
10.
Cell Rep ; 36(9): 109644, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34469735

ABSTRACT

In holometabolous insects, metamorphic timing and body size are controlled by a neuroendocrine axis composed of the ecdysone-producing prothoracic gland (PG) and its presynaptic neurons (PGNs) producing PTTH. Although PTTH/Torso signaling is considered the primary mediator of metamorphic timing, recent studies indicate that other unidentified PGN-derived factors also affect timing. Here, we demonstrate that the receptor tyrosine kinases anaplastic lymphoma kinase (Alk) and PDGF and VEGF receptor-related (Pvr), function in coordination with PTTH/Torso signaling to regulate pupariation timing and body size. Both Alk and Pvr trigger Ras/Erk signaling in the PG to upregulate expression of ecdysone biosynthetic enzymes, while Alk also suppresses autophagy by activating phosphatidylinositol 3-kinase (PI3K)/Akt. The Alk ligand Jelly belly (Jeb) is produced by the PGNs and serves as a second PGN-derived tropic factor, while Pvr activation mainly relies on autocrine signaling by PG-derived Pvf2 and Pvf3. These findings illustrate that a combination of juxtacrine and autocrine signaling regulates metamorphic timing, the defining event of holometabolous development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Endocrine Glands/enzymology , Metamorphosis, Biological , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Animals, Genetically Modified , Autocrine Communication , Body Size , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Ecdysone/metabolism , Endocrine Glands/embryology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Developmental , IMP Dehydrogenase/genetics , IMP Dehydrogenase/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Mutation , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/metabolism
11.
PLoS Genet ; 17(8): e1009738, 2021 08.
Article in English | MEDLINE | ID: mdl-34411095

ABSTRACT

Activation of Ras signaling occurs in ~30% of human cancers. However, activated Ras alone is insufficient to produce malignancy. Thus, it is imperative to identify those genes cooperating with activated Ras in driving tumoral growth. In this work, we have identified a novel EGFR inhibitor, which we have named EGFRAP, for EGFR adaptor protein. Elimination of EGFRAP potentiates activated Ras-induced overgrowth in the Drosophila wing imaginal disc. We show that EGFRAP interacts physically with the phosphorylated form of EGFR via its SH2 domain. EGFRAP is expressed at high levels in regions of maximal EGFR/Ras pathway activity, such as at the presumptive wing margin. In addition, EGFRAP expression is up-regulated in conditions of oncogenic EGFR/Ras activation. Normal and oncogenic EGFR/Ras-mediated upregulation of EGRAP levels depend on the Notch pathway. We also find that elimination of EGFRAP does not affect overall organogenesis or viability. However, simultaneous downregulation of EGFRAP and its ortholog PVRAP results in defects associated with increased EGFR function. Based on these results, we propose that EGFRAP is a new negative regulator of the EGFR/Ras pathway, which, while being required redundantly for normal morphogenesis, behaves as an important modulator of EGFR/Ras-driven tissue hyperplasia. We suggest that the ability of EGFRAP to functionally inhibit the EGFR pathway in oncogenic cells results from the activation of a feedback loop leading to increase EGFRAP expression. This could act as a surveillance mechanism to prevent excessive EGFR activity and uncontrolled cell growth.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Genes, ras/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle , Cell Proliferation/genetics , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/physiology , Imaginal Discs/metabolism , Morphogenesis , Phosphorylation , Receptors, Invertebrate Peptide/antagonists & inhibitors , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Signal Transduction/genetics , ras Proteins/genetics
12.
Elife ; 102021 06 07.
Article in English | MEDLINE | ID: mdl-34096503

ABSTRACT

RAS-like (RAL) GTPases function in Wnt signalling-dependent intestinal stem cell proliferation and regeneration. Whether RAL proteins work as canonical RAS effectors in the intestine and the mechanisms of how they contribute to tumourigenesis remain unclear. Here, we show that RAL GTPases are necessary and sufficient to activate EGFR/MAPK signalling in the intestine, via induction of EGFR internalisation. Knocking down Drosophila RalA from intestinal stem and progenitor cells leads to increased levels of plasma membrane-associated EGFR and decreased MAPK pathway activation. Importantly, in addition to influencing stem cell proliferation during damage-induced intestinal regeneration, this role of RAL GTPases impacts on EGFR-dependent tumourigenic growth in the intestine and in human mammary epithelium. However, the effect of oncogenic RAS in the intestine is independent from RAL function. Altogether, our results reveal previously unrecognised cellular and molecular contexts where RAL GTPases become essential mediators of adult tissue homeostasis and malignant transformation.


Subject(s)
Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , ErbB Receptors/metabolism , Intestinal Mucosa/metabolism , Monomeric GTP-Binding Proteins/metabolism , Receptors, Invertebrate Peptide/metabolism , Stem Cells/metabolism , ral GTP-Binding Proteins/metabolism , Animals , Animals, Genetically Modified , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Endocytosis , ErbB Receptors/genetics , Female , Humans , Hyperplasia , Intestinal Mucosa/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mammary Glands, Human/enzymology , Mammary Glands, Human/pathology , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Monomeric GTP-Binding Proteins/genetics , Receptors, Invertebrate Peptide/genetics , Signal Transduction , Stem Cells/pathology , ral GTP-Binding Proteins/genetics
13.
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
14.
Development ; 147(15)2020 08 14.
Article in English | MEDLINE | ID: mdl-32680934

ABSTRACT

To bridge the gap between qualitative and quantitative analyses of the epidermal growth factor receptor (EGFR) in tissues, we generated an sfGFP-tagged EGF receptor (EGFR-sfGFP) in Drosophila The homozygous fly appears similar to wild type with EGFR expression and activation patterns that are consistent with previous reports in the ovary, early embryo, and imaginal discs. Using ELISA, we quantified an average of 1100, 6200 and 2500 receptors per follicle cell (FC) at stages 8/9, 10 and ≥11 of oogenesis, respectively. Interestingly, the spatial localization of the EGFR to the apical side of the FCs at early stages depended on the TGFα-like ligand Gurken. At later stages, EGFR localized to basolateral positions of the FCs. Finally, we followed the endosomal localization of EGFR in the FCs. The EGFR colocalized with the late endosome, but no significant colocalization of the receptor was found with the early endosome. The EGFR-sfGFP fly is an exciting new resource for studying cellular localization and regulation of EGFR in tissues.


Subject(s)
Drosophila Proteins/metabolism , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Ovarian Follicle/metabolism , Receptors, Invertebrate Peptide/metabolism , Signal Transduction , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Endosomes/genetics , Endosomes/metabolism , Epithelial Cells/cytology , Epithelium/metabolism , ErbB Receptors/genetics , Female , Ovarian Follicle/cytology , Receptors, Invertebrate Peptide/genetics , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
15.
Dis Model Mech ; 13(6)2020 06 24.
Article in English | MEDLINE | ID: mdl-32461236

ABSTRACT

Aldosterone is produced by the mammalian adrenal cortex to modulate blood pressure and fluid balance; however, excessive, prolonged aldosterone promotes fibrosis and kidney failure. How aldosterone triggers disease may involve actions independent of its canonical mineralocorticoid receptor. Here, we present a Drosophila model of renal pathology caused by excess extracellular matrix formation, stimulated by exogenous aldosterone and by insect ecdysone. Chronic administration of aldosterone or ecdysone induces expression and accumulation of collagen-like Pericardin in adult nephrocytes - podocyte-like cells that filter circulating hemolymph. Excess Pericardin deposition disrupts nephrocyte (glomerular) filtration and causes proteinuria in Drosophila, hallmarks of mammalian kidney failure. Steroid-induced Pericardin production arises from cardiomyocytes associated with nephrocytes, potentially reflecting an analogous role of mammalian myofibroblasts in fibrotic disease. Remarkably, the canonical ecdysteroid nuclear hormone receptor, Ecdysone receptor (EcR), is not required for aldosterone or ecdysone to stimulate Pericardin production or associated renal pathology. Instead, these hormones require a cardiomyocyte-associated G-protein-coupled receptor, Dopamine-EcR (DopEcR), a membrane-associated receptor previously characterized in the fly brain to affect behavior. DopEcR in the brain is known to affect behavior through interactions with the Drosophila Epidermal growth factor receptor (Egfr), referred to as dEGFR. Here, we find that the steroids ecdysone and aldosterone require dEGFR in cardiomyocytes to induce fibrosis of the cardiac-renal system. In addition, endogenous ecdysone that becomes elevated with age is found to foster age-associated fibrosis, and to require both cardiomyocyte DopEcR and dEGFR. This Drosophila renal disease model reveals a novel signaling pathway through which steroids may modulate mammalian fibrosis through potential orthologs of DopEcR.


Subject(s)
Drosophila melanogaster/metabolism , Extracellular Matrix/metabolism , Kidney Diseases/metabolism , Kidney/metabolism , Myocytes, Cardiac/metabolism , Receptors, Steroid/metabolism , Age Factors , Aldosterone , Animals , Animals, Genetically Modified , Collagen Type IV/genetics , Collagen Type IV/metabolism , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Ecdysone , ErbB Receptors/genetics , ErbB Receptors/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Fibrosis , Kidney/pathology , Kidney Diseases/chemically induced , Kidney Diseases/genetics , Kidney Diseases/pathology , Myocytes, Cardiac/pathology , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Receptors, Steroid/genetics , Signal Transduction
16.
Curr Biol ; 30(8): 1547-1554.e4, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32220314

ABSTRACT

Understanding the mechanisms that determine final body size of animals is a central question in biology. In animals with determinate growth, such as mammals or insects, the size at which the immature organism transforms into the adult defines the final body size, as adult individuals do not grow [1]. In Drosophila, the growth period ends when the immature larva undergoes the metamorphic transition to develop the mature adult [2]. This metamorphic transition is triggered by a sharp increase of the steroid ecdysone, synthetized in the prothoracic gland (PG), that occurs at the end of the third instar larvae (L3) [3-6]. It is widely accepted that ecdysone biosynthesis in Drosophila is mainly induced by the activation of tyrosine kinase (RTK) Torso by the prothoracicotropic hormone (Ptth) produced into two pairs of neurosecretory cells that project their axons onto the PG [7, 8]. However, the fact that neither Ptth nor torso-null mutant animals arrest larval development but only present a delay in the larva-pupa transition [9-11] mandates for a reconsideration of the conventional model. Here, we show that Egfr signaling, rather than Ptth/torso, is the major contributor of ecdysone biosynthesis in Drosophila. We found that Egfr signaling is activated in the PG in an autocrine mode by the EGF ligands spitz and vein, which in turn are regulated by the levels of ecdysone. This regulatory positive feedback loop ensures the production of ecdysone to trigger metamorphosis by a progressive Egfr-dependent activation of MAPK/ERK pathway, thus determining the animal final body size.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Ecdysone/biosynthesis , ErbB Receptors/genetics , Receptors, Invertebrate Peptide/genetics , Signal Transduction/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , ErbB Receptors/metabolism , Larva/growth & development , Larva/metabolism , Pupa/growth & development , Pupa/metabolism , Receptors, Invertebrate Peptide/metabolism
17.
Cell Rep ; 30(4): 1101-1116.e5, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31995752

ABSTRACT

Although typically upregulated upon cellular stress, autophagy can also be utilized under homeostatic conditions as a quality control mechanism or in response to developmental cues. Here, we report that autophagy is required for the maintenance of somatic cyst stem cells (CySCs) in the Drosophila testis. Disruption of autophagy in CySCs and early cyst cells (CCs) by the depletion of autophagy-related (Atg) genes reduced early CC numbers and affected CC function, resembling decreased epidermal growth factor receptor (EGFR) signaling. Indeed, our data indicate that EGFR acts to stimulate autophagy to preserve early CC function, whereas target of rapamycin (TOR) negatively regulates autophagy in the differentiating CCs. Finally, we show that the EGFR-mediated stimulation of autophagy regulates lipid levels in CySCs and CCs. These results demonstrate a key role for autophagy in regulating somatic stem cell behavior and tissue homeostasis by integrating cues from both the EGFR and TOR signaling pathways to control lipid metabolism.


Subject(s)
Autophagy/genetics , Drosophila Proteins/metabolism , Drosophila/metabolism , ErbB Receptors/metabolism , Germ Cells/metabolism , Lipid Metabolism/genetics , Receptors, Invertebrate Peptide/metabolism , Stem Cells/metabolism , Animals , Animals, Genetically Modified , Autophagosomes/metabolism , Cell Differentiation/genetics , Drosophila Proteins/genetics , ErbB Receptors/genetics , Gene Knockdown Techniques , Germ Cells/growth & development , Homeostasis , MAP Kinase Signaling System/genetics , Male , RNA Interference , Receptors, Invertebrate Peptide/genetics , TOR Serine-Threonine Kinases/metabolism , Testis/cytology , Testis/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
18.
Annu Rev Genet ; 53: 1-18, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31794267

ABSTRACT

In Drosophila development, the axes of the egg and future embryo are established during oogenesis. To learn about the underlying genetic and molecular pathways that lead to axis formation, I conducted a large-scale genetic screen at the beginning of my independent career. This led to the eventual understanding that both anterior-posterior and dorsal-ventral pattern information is transmitted from the oocyte to the surrounding follicle cells and in turn from the follicle cells back to the oocyte. How I came to conduct this screen and what further insights were gained by studying the mutants isolated in the screen are the topics of this autobiographical article.


Subject(s)
Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Genetics/history , Ovum/physiology , Animals , Body Patterning/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Developmental , History, 20th Century , History, 21st Century , Male , Oocytes/physiology , Ovary/growth & development , Ovary/physiology , Receptors, Invertebrate Peptide/genetics , Receptors, Invertebrate Peptide/metabolism , Sex Determination Analysis , Sex Determination Processes , United States
19.
Sci Rep ; 9(1): 18628, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31819141

ABSTRACT

In all metazoans, a small number of evolutionarily conserved signaling pathways are reiteratively used during development to orchestrate critical patterning and morphogenetic processes. Among these, Notch (N) signaling is essential for most aspects of tissue patterning where it mediates the communication between adjacent cells to control cell fate specification. In Drosophila, Notch signaling is required for several features of eye development, including the R3/R4 cell fate choice and R7 specification. Here we show that hypomorphic alleles of Notch, belonging to the Nfacet class, reveal a novel phenotype: while photoreceptor specification in the mutant ommatidia is largely normal, defects are observed in ommatidial rotation (OR), a planar cell polarity (PCP)-mediated cell motility process. We demonstrate that during OR Notch signaling is specifically required in the R4 photoreceptor to upregulate the transcription of argos (aos), an inhibitory ligand to the epidermal growth factor receptor (EGFR), to fine-tune the activity of EGFR signaling. Consistently, the loss-of-function defects of Nfacet alleles and EGFR-signaling pathway mutants are largely indistinguishable. A Notch-regulated aos enhancer confers R4 specific expression arguing that aos is directly regulated by Notch signaling in this context via Su(H)-Mam-dependent transcription.


Subject(s)
Drosophila Proteins/genetics , ErbB Receptors/genetics , Eye Proteins/genetics , Nerve Tissue Proteins/genetics , Receptors, Invertebrate Peptide/genetics , Receptors, Notch/genetics , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Polarity/genetics , Drosophila melanogaster/genetics , Eye/metabolism , Frizzled Receptors/genetics , Morphogenesis/genetics , Photoreceptor Cells, Invertebrate/metabolism
20.
Sci Rep ; 9(1): 19695, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31873089

ABSTRACT

Mitochondria are essential organelles that have recently emerged as hubs for several metabolic and signaling pathways in the cell. Mitochondrial morphology is regulated by constant fusion and fission events to maintain a functional mitochondrial network and to remodel the mitochondrial network in response to external stimuli. Although the role of mitochondria in later stages of spermatogenesis has been investigated in depth, the role of mitochondrial dynamics in regulating early germ cell behavior is relatively less-well understood. We previously demonstrated that mitochondrial fusion is required for germline stem cell (GSC) maintenance in the Drosophila testis. Here, we show that mitochondrial fission is also important for regulating the maintenance of early germ cells in larval testes. Inhibition of Drp1 in early germ cells resulted in the loss of GSCs and spermatogonia due to the accumulation of reactive oxygen species (ROS) and activation of the EGFR pathway in adjacent somatic cyst cells. EGFR activation contributed to premature germ cell differentiation. Our data provide insights into how mitochondrial dynamics can impact germ cell maintenance and differentiation via distinct mechanisms throughout development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , ErbB Receptors/metabolism , Mitochondrial Dynamics/physiology , Receptors, Invertebrate Peptide/metabolism , Spermatozoa/metabolism , Testis/cytology , Testis/metabolism , Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/metabolism , Animals , Animals, Genetically Modified , Cell Differentiation , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , GTP-Binding Proteins/deficiency , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Larva/cytology , Larva/metabolism , Male , Reactive Oxygen Species/metabolism , Receptors, Invertebrate Peptide/genetics , Signal Transduction , Spermatogenesis/physiology , Spermatogonia/cytology , Spermatogonia/metabolism , Spermatozoa/cytology
SELECTION OF CITATIONS
SEARCH DETAIL