Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 312
Filter
1.
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
2.
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
3.
J Cell Biochem ; 125(6): e30545, 2024 06.
Article in English | MEDLINE | ID: mdl-38436545

ABSTRACT

To maintain the integrity of the adult gut, the proliferation and differentiation of stem cells must be strictly controlled. Several signaling pathways control the proliferation and differentiation of Drosophila intestinal epithelial cells. Although the modulatory effects of insulin pathway components on cell proliferation have been characterized, their specific role in which cell type and how these components interact with other regulatory signaling pathways remain largely unclear. In this study, we found that InR/Pi3K has major functions in enteroblasts (EBs) that were not previously described. The absence of InR/Pi3K in progenitors leads to a decrease in the number of EBs, while it has no significant effect on intestinal stem cells (ISCs). In addition, we found that InR/Pi3K regulates Notch activity in ISCs and EBs in an opposite way. This is also the reason for the decrease in EB. On the one hand, aberrantly low levels of Notch signaling in ISCs inhibit their proper differentiation into EBs; on the other hand, the higher Notch levels in EBs promote their excessive differentiation into enterocytes (ECs), leading to marked increases in abnormal ECs and decreased proliferation. Moreover, we found that Upd/JAK/STAT signaling acts as an effector or modifier of InR/Pi3K function in the midgut and cooperates with EGFR signaling to regulate cell proliferation. Altogether, our results demonstrate that InR and Pi3K are essential for coordinating stem cell differentiation and proliferation to maintain intestinal homeostasis.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Phosphatidylinositol 3-Kinases , Signal Transduction , Animals , Cell Differentiation , Cell Proliferation , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Enterocytes/metabolism , Enterocytes/cytology , ErbB Receptors/metabolism , Homeostasis , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Intestines/cytology , Phosphatidylinositol 3-Kinases/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Invertebrate Peptide , Receptors, Notch/metabolism , STAT Transcription Factors/metabolism , Stem Cells/metabolism , Stem Cells/cytology
4.
Dev Cell ; 59(8): 1075-1090.e6, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38521056

ABSTRACT

The Drosophila lymph gland houses blood progenitors that give rise to myeloid-like blood cells. Initially, blood progenitors proliferate, but later, they become quiescent to maintain multipotency before differentiation. Despite the identification of various factors involved in multipotency maintenance, the cellular mechanism controlling blood progenitor quiescence remains elusive. Here, we identify the expression of nitric oxide synthase in blood progenitors, generating nitric oxide for post-translational S-nitrosylation of protein cysteine residues. S-nitrosylation activates the Ire1-Xbp1-mediated unfolded protein response, leading to G2 cell-cycle arrest. Specifically, we identify the epidermal growth factor receptor as a target of S-nitrosylation, resulting in its retention within the endoplasmic reticulum and blockade of its receptor function. Overall, our findings highlight developmentally programmed S-nitrosylation as a critical mechanism that induces protein quality control in blood progenitors, maintaining their undifferentiated state by inhibiting cell-cycle progression and rendering them unresponsive to paracrine factors.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Endoribonucleases , Hematopoietic Stem Cells , Receptors, Invertebrate Peptide , Unfolded Protein Response , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Drosophila melanogaster/metabolism , Nitric Oxide/metabolism , ErbB Receptors/metabolism , Cell Differentiation , Endoplasmic Reticulum/metabolism , Nitric Oxide Synthase/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Signal Transduction
5.
Cells ; 12(4)2023 02 19.
Article in English | MEDLINE | ID: mdl-36831328

ABSTRACT

The Drosophila lymph gland is an ideal model for studying hematopoiesis, and unraveling the mechanisms of Drosophila hematopoiesis can improve our understanding of the pathogenesis of human hematopoietic malignancies. Bone morphogenetic protein (BMP) signaling is involved in a variety of biological processes and is highly conserved between Drosophila and mammals. Decapentaplegic (Dpp)/BMP signaling is known to limit posterior signaling center (PSC) cell proliferation by repressing the protooncogene dmyc. However, the role of two other TGF-ß family ligands, Glass bottom boat (Gbb) and Screw (Scw), in Drosophila hematopoiesis is currently largely unknown. Here, we showed that the loss of Gbb in the cortical zone (CZ) induced lamellocyte differentiation by overactivation of the EGFR and JNK pathways and caused excessive differentiation of plasmatocytes, mainly by the hyperactivation of EGFR. Furthermore, we found that Gbb was also required for preventing the hyperproliferation of the lymph glands by inhibiting the overactivation of the Epidermal Growth Factor Receptor (EGFR) and c-Jun N-terminal Kinase (JNK) pathways. These results further advance our understanding of the roles of Gbb protein and the BMP signaling in Drosophila hematopoiesis and the regulatory relationship between the BMP, EGFR, and JNK pathways in the proliferation and differentiation of lymph gland hemocytes.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Humans , Drosophila/metabolism , Drosophila Proteins/metabolism , Signal Transduction/physiology , Hematopoiesis , ErbB Receptors/metabolism , Cell Proliferation , Mammals/metabolism , Receptors, Invertebrate Peptide
6.
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
7.
Curr Biol ; 33(3): 517-532.e5, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36640763

ABSTRACT

The development of neuronal connectivity requires stabilization of dynamic axonal branches at sites of synapse formation. Models that explain how axonal branching is coupled to synaptogenesis postulate molecular regulators acting in a spatiotemporally restricted fashion to ensure branching toward future synaptic partners while also stabilizing the emerging synaptic contacts between such partners. We investigated this question using neuronal circuit development in the Drosophila brain as a model system. We report that epidermal growth factor receptor (EGFR) activity is required in presynaptic axonal branches during two distinct temporal intervals to regulate circuit wiring in the developing Drosophila visual system. EGFR is required early to regulate primary axonal branching. EGFR activity is then independently required at a later stage to prevent degradation of the synaptic active zone protein Bruchpilot (Brp). Inactivation of EGFR results in a local increase of autophagy in presynaptic branches and the translocation of active zone proteins into autophagic vesicles. The protection of synaptic material during this later interval of wiring ensures the stabilization of terminal branches, circuit connectivity, and appropriate visual behavior. Phenotypes of EGFR inactivation can be rescued by increasing Brp levels or downregulating autophagy. In summary, we identify a temporally restricted molecular mechanism required for coupling axonal branching and synaptic stabilization that contributes to the emergence of neuronal wiring specificity.


Subject(s)
Drosophila Proteins , Animals , Drosophila Proteins/metabolism , Axons/physiology , Drosophila/genetics , ErbB Receptors/metabolism , Autophagy , Synapses/physiology , Receptors, Invertebrate Peptide/metabolism
8.
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
9.
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
10.
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
11.
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
12.
Cell Prolif ; 55(1): e13173, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34952996

ABSTRACT

OBJECTIVES: Adult stem cells uphold a delicate balance between quiescent and active states, which is crucial for tissue homeostasis. Whereas many signalling pathways that regulate epithelial stem cells have been reported, many regulators remain unidentified. MATERIALS AND METHODS: Flies were used to generate tissue-specific gene knockdown and gene knockout. qRT-PCR was used to assess the relative mRNA levels. Immunofluorescence was used to determine protein localization and expression patterns. Clonal analyses were used to observe the phenotype. RNA-seq was used to screen downstream mechanisms. RESULTS: Here, we report a member of the chloride channel family, ClC-c, which is specifically expressed in Drosophila intestinal stem/progenitor cells and regulates intestinal stem cell (ISC) proliferation under physiological conditions and upon tissue damage. Mechanistically, we found that the ISC loss induced by the depletion of ClC-c in intestinal stem/progenitor cells is due to inhibition of the EGFR signalling pathway. CONCLUSION: Our findings reveal an ISC-specific function of ClC-c in regulating stem cell maintenance and proliferation, thereby providing new insights into the functional links among the chloride channel family, ISC proliferation and tissue homeostasis.


Subject(s)
Chloride Channels/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , ErbB Receptors/metabolism , Intestines/cytology , Receptors, Invertebrate Peptide/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Animals , Apoptosis/genetics , Base Sequence , Cell Proliferation , Down-Regulation/genetics , Endosomes/metabolism , Intestinal Mucosa/cytology , Necrosis , Regeneration , rab5 GTP-Binding Proteins/metabolism
13.
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
14.
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
15.
Dev Biol ; 478: 173-182, 2021 10.
Article in English | MEDLINE | ID: mdl-34245727

ABSTRACT

A complex network of transcription factor interactions propagates across the larval eye disc to establish columns of evenly-spaced R8 precursor cells, the founding cells of Drosophila ommatidia. After the recruitment of additional photoreceptors to each ommatidium, the surrounding cells are organized into their stereotypical pattern during pupal development. These support cells - comprised of pigment and cone cells - are patterned to encapsulate the photoreceptors and separate ommatidia with an hexagonal honeycomb lattice. Since the proteins and processes essential for correct eye patterning are conserved, elucidating how these function and change during Drosophila eye patterning can substantially advance our understanding of transcription factor and signaling networks, cytoskeletal structures, adhesion complexes, and the biophysical properties of complex tissues during their morphogenesis. Our understanding of many of these aspects of Drosophila eye patterning is largely descriptive. Many important questions, especially relating to the regulation and integration of cellular events, remain.


Subject(s)
Compound Eye, Arthropod/growth & development , Drosophila/growth & development , Photoreceptor Cells, Invertebrate/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Body Patterning , Compound Eye, Arthropod/cytology , Computer Simulation , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Larva/growth & development , Morphogenesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Photoreceptor Cells, Invertebrate/cytology , Pupa/growth & development , Receptors, Invertebrate Peptide/metabolism , Signal Transduction
16.
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
17.
Dev Biol ; 477: 133-144, 2021 09.
Article in English | MEDLINE | ID: mdl-34044021

ABSTRACT

The Drosophila testis is a model organism stem cell niche in which two stem cell populations coordinate together to produce sperm; thus, these stem cells must be balanced in the niche. Merlin, a tumor-suppressor and human disease gene required for contact inhibition of proliferation, is known to limit the proliferation of the somatic cyst stem cells in the testis niche. Expanded encodes a protein that is structurally similar to Merlin in Drosophila, and is semi-redundant with Merlin in multiple tissues. We found that expanded depletion caused similar cyst lineage cell over-proliferation as observed with Merlin, and double mutants showed more severe phenotypes than either gene individually. Thus, these genes have partially redundant functions in the cyst lineage cells of this niche. We also expressed non-phosphorylatable constitutively "tumor suppressing" alleles of Merlin in cyst lineage cells, and surprisingly, we observed a similar cyst lineage over-proliferation phenotype. Merlin is known to impact multiple different signaling pathways to exert its effect on proliferation. We found that the Merlin loss of function phenotype was associated with an increase in MAPK/ERK signaling, consistent with Merlin's established role in transmembrane receptor inhibition. Constitutive Merlin displayed a reduction in both MAPK/ERK signaling and PI3K/Tor signaling. PI3K/Tor signaling is required for cyst cell differentiation, and inhibition of this pathway by Merlin activation phenocopied the Tor cyst lineage loss of function phenotype. Thus, Merlin impacts and integrates the activity of multiple signaling pathways in the testis niche. The ability of Merlin to dynamically change its activity via phosphorylation in response to local contact cues provides an intriguing mechanism whereby the signaling pathways that control these stem cells might be dynamically regulated in response to the division of a neighboring germ cell.


Subject(s)
Adult Stem Cells/physiology , Cell Proliferation/physiology , Drosophila Proteins/physiology , Drosophila/cytology , Membrane Proteins/physiology , Neurofibromin 2/physiology , Signal Transduction , Testis/cytology , Animals , Cell Lineage , Drosophila/embryology , Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Male , Models, Biological , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Invertebrate Peptide/metabolism , Testis/embryology
18.
Cell Death Dis ; 12(5): 491, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990549

ABSTRACT

Spermatogonia transit-amplifying (TA) divisions are crucial for the differentiation of germline stem cell daughters. However, the underlying mechanism is largely unknown. In the present study, we demonstrated that CG6015 was essential for spermatogonia TA-divisions and elongated spermatozoon development in Drosophila melanogaster. Spermatogonia deficient in CG6015 inhibited germline differentiation leading to the accumulation of undifferentiated cell populations. Transcriptome profiling using RNA sequencing indicated that CG6015 was involved in spermatogenesis, spermatid differentiation, and metabolic processes. Gene Set Enrichment Analysis (GSEA) revealed the relationship between CG6015 and the epidermal growth factor receptor (EGFR) signaling pathway. Unexpectedly, we discovered that phosphorylated extracellular regulated kinase (dpERK) signals were activated in germline stem cell (GSC)-like cells after reduction of CG6015 in spermatogonia. Moreover, Downstream of raf1 (Dsor1), a key downstream target of EGFR, mimicked the phenotype of CG6015, and germline dpERK signals were activated in spermatogonia of Dsor1 RNAi testes. Together, these findings revealed a potential regulatory mechanism of CG6015 via EGFR signaling during spermatogonia TA-divisions in Drosophila testes.


Subject(s)
Drosophila Proteins/metabolism , ErbB Receptors/metabolism , Receptors, Invertebrate Peptide/metabolism , Spermatogonia/physiology , Testis/metabolism , Animals , Cell Differentiation/physiology , Drosophila , Male
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...