Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
FEBS Lett ; 598(4): 379-389, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38351618

ABSTRACT

Multicellular communities have an intrinsic mechanism that optimizes their structure and function via cell-cell communication. One of the driving forces for such self-organization of the multicellular system is cell competition, the elimination of viable unfit or deleterious cells via cell-cell interaction. Studies in Drosophila and mammals have identified multiple mechanisms of cell competition caused by different types of mutations or cellular changes. Intriguingly, recent studies have found that different types of "losers" of cell competition commonly show reduced protein synthesis. In Drosophila, the reduction in protein synthesis levels in loser cells is caused by phosphorylation of the translation initiation factor eIF2α via a bZip transcription factor Xrp1. Given that a variety of cellular stresses converge on eIF2α phosphorylation and thus global inhibition of protein synthesis, cell competition may be a machinery that optimizes multicellular fitness by removing stressed cells. In this review, we summarize and discuss emerging signaling mechanisms and critical unsolved questions, as well as the role of protein synthesis in cell competition.


Subject(s)
Drosophila Proteins , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Cell Competition , Signal Transduction , Drosophila/metabolism , Cell Communication , Mammals/metabolism , DNA-Binding Proteins/metabolism
2.
Cell Struct Funct ; 49(1): 11-20, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38199250

ABSTRACT

The ribosome is a molecular machine essential for protein synthesis, which is composed of approximately 80 different ribosomal proteins (Rps). Studies in yeast and cell culture systems have revealed that the intracellular level of Rps is finely regulated by negative feedback mechanisms or ubiquitin-proteasome system, which prevents over- or under-abundance of Rps in the cell. However, in vivo evidence for the homeostatic regulation of intracellular Rp levels has been poor. Here, using Drosophila genetics, we show that intracellular Rp levels are regulated by proteasomal degradation of excess Rps that are not incorporated into the ribosome. By establishing an EGFP-fused Rp gene system that can monitor endogenously expressed Rp levels, we found that endogenously expressed EGFP-RpS20 or -RpL5 is eliminated from the cell when RpS20 or RpL5 is exogenously expressed. Notably, the level of endogenously expressed Hsp83, a housekeeping gene, was not affected by exogenous expression of Hsp83, suggesting that the strict negative regulation of excess protein is specific for intracellular Rps. Further analyses revealed that the maintenance of cellular Rp levels is not regulated at the transcriptional level but by proteasomal degradation of excess free Rps as a protein quality control mechanism. Our observations provide not only the in vivo evidence for the homeostatic regulation of Rp levels but also a novel genetic strategy to study in vivo regulation of intracellular Rp levels and its role in tissue homeostasis via cell competition.Key words: ribosomal protein, proteasomal degradation, Drosophila.


Subject(s)
Drosophila , Ribosomal Proteins , Animals , Drosophila/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ribosomes/metabolism , Protein Biosynthesis , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Saccharomyces cerevisiae/metabolism
3.
Fly (Austin) ; 17(1): 2173997, 2023 12.
Article in English | MEDLINE | ID: mdl-36876479
4.
PLoS Genet ; 19(3): e1010684, 2023 03.
Article in English | MEDLINE | ID: mdl-36972315

ABSTRACT

The function of the stem cell system is supported by a stereotypical shape of the niche structure. In Drosophila ovarian germarium, somatic cap cells form a dish-like niche structure that allows only two or three germ-line stem cells (GSCs) reside in the niche. Despite extensive studies on the mechanism of stem cell maintenance, the mechanisms of how the dish-like niche structure is shaped and how this structure contributes to the stem cell system have been elusive. Here, we show that a transmembrane protein Stranded at second (Sas) and its receptor Protein tyrosine phosphatase 10D (Ptp10D), effectors of axon guidance and cell competition via epidermal growth factor receptor (Egfr) inhibition, shape the dish-like niche structure by facilitating c-Jun N-terminal kinase (JNK)-mediated apoptosis. Loss of Sas or Ptp10D in gonadal apical cells, but not in GSCs or cap cells, during the pre-pupal stage results in abnormal shaping of the niche structure in the adult, which allows excessive, four to six GSCs reside in the niche. Mechanistically, loss of Sas-Ptp10D elevates Egfr signaling in the gonadal apical cells, thereby suppressing their naturally-occurring JNK-mediated apoptosis that is essential for the shaping of the dish-like niche structure by neighboring cap cells. Notably, the abnormal niche shape and resulting excessive GSCs lead to diminished egg production. Our data propose a concept that the stereotypical shaping of the niche structure optimizes the stem cell system, thereby maximizing the reproductive capacity.


Subject(s)
Drosophila Proteins , Animals , Apoptosis/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Germ Cells/metabolism , Phosphoric Monoester Hydrolases/metabolism , Stem Cell Niche/genetics , Proto-Oncogene Proteins c-jun/metabolism
5.
Fly (Austin) ; 16(1): 367-381, 2022 12.
Article in English | MEDLINE | ID: mdl-36413374

ABSTRACT

Cell-cell interactions within tumour microenvironment play crucial roles in tumorigenesis. Genetic mosaic techniques available in Drosophila have provided a powerful platform to study the basic principles of tumour growth and progression via cell-cell communications. This led to the identification of oncogenic cell-cell interactions triggered by endocytic dysregulation, mitochondrial dysfunction, cell polarity defects, or Src activation in Drosophila imaginal epithelia. Such oncogenic cooperations can be caused by interactions among epithelial cells, mesenchymal cells, and immune cells. Moreover, microenvironmental factors such as nutrients, local tissue structures, and endogenous growth signalling activities critically affect tumorigenesis. Dissecting various types of oncogenic cell-cell interactions at the single-cell level in Drosophila will greatly increase our understanding of how tumours progress in living animals.


Subject(s)
Cell Communication , Drosophila , Animals , Carcinogenesis , Signal Transduction , Cell Polarity , Tumor Microenvironment
6.
Elife ; 112022 Oct 12.
Article in English | MEDLINE | ID: mdl-36222503

ABSTRACT

JNK signaling plays a critical role in both tumor promotion and tumor suppression. Here, we identified clustered microRNAs (miRNAs) miR-306 and miR-79 as novel tumor-suppressor miRNAs that specifically eliminate JNK-activated tumors in Drosophila. While showing only a slight effect on normal tissue growth, miR-306 and miR-79 strongly suppressed growth of multiple tumor models, including malignant tumors caused by Ras activation and cell polarity defects. Mechanistically, these miRNAs commonly target the mRNA of an E3 ubiquitin ligase ring finger protein 146 (RNF146). We found that RNF146 promotes degradation of tankyrase (Tnks), an ADP-ribose polymerase that promotes JNK activation in a noncanonical manner. Thus, downregulation of RNF146 by miR-306 and miR-79 leads to hyper-enhancement of JNK activation. Our data show that, while JNK activity is essential for tumor growth, elevation of miR-306 or miR-79 overactivate JNK signaling to the lethal level via noncanonical JNK pathway and thus eliminate tumors, providing a new miRNA-based strategy against cancer.


Subject(s)
MicroRNAs , Neoplasms , Humans , MicroRNAs/genetics , MAP Kinase Signaling System , Genes, Tumor Suppressor , Neoplasms/genetics , Adenosine Diphosphate Ribose
7.
Curr Biol ; 32(5): 1064-1076.e4, 2022 03 14.
Article in English | MEDLINE | ID: mdl-35134324

ABSTRACT

Mutations in the tumor-suppressor Hippo pathway lead to activation of the transcriptional coactivator Yorkie (Yki), which enhances cell proliferation autonomously and causes cell death non-autonomously. While Yki-induced cell proliferation has extensively been studied, the mechanism by which Yki causes cell death in nearby wild-type cells, a phenomenon called supercompetition, and its role in tumorigenesis remained unknown. Here, we show that Yki-induced supercompetition is essential for tumorigenesis and is driven by non-autonomous induction of autophagy. Clones of cells mutant for a Hippo pathway component fat activate Yki and cause autonomous tumorigenesis and non-autonomous cell death in Drosophila eye-antennal discs. Through a genetic screen in Drosophila, we find that mutations in autophagy-related genes or NF-κB genes in surrounding wild-type cells block both fat-induced tumorigenesis and supercompetition. Mechanistically, fat mutant cells upregulate Yki-target microRNA bantam, which elevates protein synthesis levels via activation of TOR signaling. This induces elevation of autophagy in neighboring wild-type cells, which leads to downregulation of IκB Cactus and thus causes NF-κB-mediated induction of the cell death gene hid. Crucially, upregulation of bantam is sufficient to make cells to be supercompetitors and downregulation of endogenous bantam is sufficient for cells to become losers of cell competition. Our data indicate that cells with elevated Yki-bantam signaling cause tumorigenesis by non-autonomous induction of autophagy that kills neighboring wild-type cells.


Subject(s)
Autophagy , Cell Competition , Drosophila Proteins , MicroRNAs , YAP-Signaling Proteins , Animals , Autophagy/genetics , Carcinogenesis , Cell Competition/genetics , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Hippo Signaling Pathway/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , YAP-Signaling Proteins/genetics , YAP-Signaling Proteins/metabolism
8.
PLoS Genet ; 17(12): e1009958, 2021 12.
Article in English | MEDLINE | ID: mdl-34871307

ABSTRACT

Cell competition is a context-dependent cell elimination via cell-cell interaction whereby unfit cells ('losers') are eliminated from the tissue when confronted with fitter cells ('winners'). Despite extensive studies, the mechanism that drives loser's death and its physiological triggers remained elusive. Here, through a genetic screen in Drosophila, we find that endoplasmic reticulum (ER) stress causes cell competition. Mechanistically, ER stress upregulates the bZIP transcription factor Xrp1, which promotes phosphorylation of the eukaryotic translation initiation factor eIF2α via the kinase PERK, leading to cell elimination. Surprisingly, our genetic data show that different cell competition triggers such as ribosomal protein mutations or RNA helicase Hel25E mutations converge on upregulation of Xrp1, which leads to phosphorylation of eIF2α and thus causes reduction in global protein synthesis and apoptosis when confronted with wild-type cells. These findings not only uncover a core pathway of cell competition but also open the way to understanding the physiological triggers of cell competition.


Subject(s)
Cell Competition/genetics , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Eukaryotic Initiation Factor-2/genetics , eIF-2 Kinase/genetics , Animals , Apoptosis/genetics , Drosophila melanogaster/genetics , Endoplasmic Reticulum , Endoplasmic Reticulum Stress/genetics , Phosphorylation , Signal Transduction/genetics , Transcriptional Activation/genetics
9.
PLoS Genet ; 17(11): e1009893, 2021 11.
Article in English | MEDLINE | ID: mdl-34780467

ABSTRACT

Identifying a common oncogenesis pathway among tumors with different oncogenic mutations is critical for developing anti-cancer strategies. Here, we performed transcriptome analyses on two different models of Drosophila malignant tumors caused by Ras activation with cell polarity defects (RasV12/scrib-/-) or by microRNA bantam overexpression with endocytic defects (bantam/rab5-/-), followed by an RNAi screen for genes commonly essential for tumor growth and malignancy. We identified that Juvenile hormone Inducible-21 (JhI-21), a Drosophila homolog of the L-amino acid transporter 1 (LAT1), is upregulated in these malignant tumors with different oncogenic mutations and knocking down of JhI-21 strongly blocked their growth and invasion. JhI-21 expression was induced by simultaneous activation of c-Jun N-terminal kinase (JNK) and Yorkie (Yki) in these tumors and thereby contributed to tumor growth and progression by activating the mTOR-S6 pathway. Pharmacological inhibition of LAT1 activity in Drosophila larvae significantly suppressed growth of RasV12/scrib-/- tumors. Intriguingly, LAT1 inhibitory drugs did not suppress growth of bantam/rab5-/- tumors and overexpression of bantam rendered RasV12/scrib-/- tumors unresponsive to LAT1 inhibitors. Further analyses with RNA sequencing of bantam-expressing clones followed by an RNAi screen suggested that bantam induces drug resistance against LAT1 inhibitors via downregulation of the TMEM135-like gene CG31157. Our observations unveil an evolutionarily conserved role of LAT1 induction in driving Drosophila tumor malignancy and provide a powerful genetic model for studying cancer progression and drug resistance.


Subject(s)
Amino Acid Transport Systems/metabolism , Carcinogenesis/genetics , Carcinogenesis/pathology , Drosophila Proteins/genetics , Drug Resistance, Neoplasm , MAP Kinase Kinase 4/metabolism , YAP-Signaling Proteins/metabolism , Amino Acid Transport Systems/antagonists & inhibitors , Amino Acid Transport Systems/genetics , Animals , Drosophila , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , MAP Kinase Kinase 4/genetics , MicroRNAs/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , RNA Interference , Signal Transduction , Up-Regulation , YAP-Signaling Proteins/genetics
10.
Dev Cell ; 56(15): 2223-2236.e5, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34324859

ABSTRACT

Cancer tissue often comprises multiple tumor clones with distinct oncogenic alterations such as Ras or Src activation, yet the mechanism by which tumor heterogeneity drives cancer progression remains elusive. Here, we show in Drosophila imaginal epithelium that clones of Ras- or Src-activated benign tumors interact with each other to mutually promote tumor malignancy. Mechanistically, Ras-activated cells upregulate the cell-surface ligand Delta while Src-activated cells upregulate its receptor Notch, leading to Notch activation in Src cells. Elevated Notch signaling induces the transcriptional repressor Zfh1/ZEB1, which downregulates E-cadherin and cell death gene hid, leading to Src-activated invasive tumors. Simultaneously, Notch activation in Src cells upregulates the cytokine Unpaired/IL-6, which activates JAK-STAT signaling in neighboring Ras cells. Elevated JAK-STAT signaling upregulates the BTB-zinc-finger protein Chinmo, which downregulates E-cadherin and thus generates Ras-activated invasive tumors. Our findings provide a mechanistic explanation for how tumor heterogeneity triggers tumor progression via cell-cell interactions.


Subject(s)
Neoplasms/metabolism , Oncogene Protein pp60(v-src)/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cadherins/metabolism , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Epithelium/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/genetics , Genes, ras/physiology , Imaginal Discs/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Protein pp60(v-src)/physiology , Proto-Oncogene Proteins p21(ras)/physiology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Zinc Fingers
11.
Development ; 148(14)2021 07 15.
Article in English | MEDLINE | ID: mdl-34313318

ABSTRACT

Heterozygosity of ribosomal protein genes causes a variety of developmental abnormalities in humans, which are collectively known as ribosomopathies, yet the underlying mechanisms remain elusive. Here, we analyzed Drosophila Minute (M)/+ mutants, a group of mutants heterozygous for ribosomal protein genes that exhibit a characteristic thin-bristle phenotype. We found that, although M/+ flies develop essentially normal wings, simultaneous deletion of one copy of the Hippo pathway effector yki resulted in severe wing growth defects. These defects were caused by JNK-mediated cell death in the wing pouch via Eiger/TNF signaling. The JNK activation in M/+, yki/+ wing discs required the caspase Dronc, which is normally blocked by DIAP1. Notably, heterozygosity of yki reduced DIAP1 expression in the wing pouch, leading to elevation of Dronc activity. Dronc and JNK formed a positive-feedback loop that amplifies Dronc activation, leading to apoptosis. Our observations suggest a mechanism of robust tissue growth whereby tissues with reduced ribosomal protein prevent ectopic apoptosis via Yki activity.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Animals , Apoptosis , Cell Death , Down-Regulation , Drosophila/genetics , Female , Gene Expression Regulation, Developmental , Inhibitor of Apoptosis Proteins/metabolism , Nuclear Proteins/genetics , Signal Transduction , Trans-Activators/genetics , Wings, Animal/anatomy & histology , Wings, Animal/metabolism , YAP-Signaling Proteins
12.
Sci Signal ; 14(685)2021 06 01.
Article in English | MEDLINE | ID: mdl-34074704

ABSTRACT

The activation of Ras signaling is a major early event of oncogenesis in many contexts, yet paradoxically, Ras signaling induces cellular senescence, which prevents tumorigenesis. Thus, Ras-activated cells must overcome senescence to develop into cancer. Through a genetic screen in Drosophila melanogaster, we found that the ETS family transcriptional activator Pointed (Pnt) was necessary and sufficient to trigger cellular senescence upon Ras activation and blocked Ras-induced tumor growth in eye-antennal discs. Through analyses of mosaic discs using various genetic tools, we identified a mechanism of tumor progression in which loss of cell polarity, a common driver of epithelial oncogenesis, abrogated Ras-induced cellular senescence through microRNA-mediated inhibition of Pnt. Mechanistically, polarity defects in Ras-activated cells caused activation of the Hippo effector Yorkie (Yki), which induced the expression of the microRNA bantam bantam-mediated repression of the E3 ligase-associated protein Tribbles (Trbl) relieved Ras- and Akt-dependent inhibition of the transcription factor FoxO. The restoration of FoxO activity in Ras-activated cells induced the expression of the microRNAs miR-9c and miR-79, which led to reduced pnt expression, thereby abrogating cellular senescence and promoting tumor progression. Our findings provide a mechanistic explanation for how Ras-activated tumors progress toward malignancy by overcoming cellular senescence.


Subject(s)
Cellular Senescence , Drosophila Proteins/metabolism , MicroRNAs , YAP-Signaling Proteins/metabolism , Animals , Carcinogenesis , Cell Proliferation , DNA-Binding Proteins , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , MicroRNAs/genetics , Nerve Tissue Proteins , Nuclear Proteins , Proto-Oncogene Proteins , Trans-Activators , Transcription Factors
13.
PLoS Genet ; 17(1): e1009300, 2021 01.
Article in English | MEDLINE | ID: mdl-33507966

ABSTRACT

Highly reproducible tissue development is achieved by robust, time-dependent coordination of cell proliferation and cell death. To study the mechanisms underlying robust tissue growth, we analyzed the developmental process of wing imaginal discs in Drosophila Minute mutants, a series of heterozygous mutants for a ribosomal protein gene. Minute animals show significant developmental delay during the larval period but develop into essentially normal flies, suggesting there exists a mechanism ensuring robust tissue growth during abnormally prolonged developmental time. Surprisingly, we found that both cell death and compensatory cell proliferation were dramatically increased in developing wing pouches of Minute animals. Blocking the cell-turnover by inhibiting cell death resulted in morphological defects, indicating the essential role of cell-turnover in Minute wing morphogenesis. Our analyses showed that Minute wing discs elevate Wg expression and JNK-mediated Dilp8 expression that causes developmental delay, both of which are necessary for the induction of cell-turnover. Furthermore, forced increase in Wg expression together with developmental delay caused by ecdysone depletion induced cell-turnover in the wing pouches of non-Minute animals. Our findings suggest a novel paradigm for robust coordination of tissue growth by cell-turnover, which is induced when developmental time axis is distorted.


Subject(s)
Drosophila Proteins/genetics , Imaginal Discs/growth & development , Intercellular Signaling Peptides and Proteins/genetics , Ribosomal Proteins/genetics , Wnt1 Protein/genetics , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ecdysone/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Imaginal Discs/metabolism , Larva/genetics , Larva/growth & development , Metamorphosis, Biological/genetics , Organogenesis/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Wings, Animal/growth & development , Wings, Animal/metabolism
14.
Cancer Sci ; 111(10): 3409-3415, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32677169

ABSTRACT

Oncogenic mutations often trigger antitumor cellular response such as induction of apoptosis or cellular senescence. Studies in the last decade have identified the presence of the third guardian against mutation-induced tumorigenesis, namely "cell competition." Cell competition is a context-dependent cell elimination whereby cells with higher fitness eliminate neighboring cells with lower fitness by inducing cell death. While oncogene-induced apoptosis or oncogene-induced senescence acts as a cell-autonomous tumor suppressor, cell competition protects the tissue from tumorigenesis via cell-cell communication. For instance, in Drosophila epithelium, oncogenic cells with cell polarity mutations overproliferate and develop into tumors on their own but are eliminated from the tissue when surrounded by wild-type cells. Genetic studies in flies have unraveled that such tumor-suppressive cell competition is regulated by at least three mechanisms: direct cell-cell interaction between polarity-deficient cells and wild-type cells, secreted factors from epithelial cells, and systemic factors from distant organs. Molecular manipulation of tumor-suppressive cell competition could provide a novel therapeutic strategy against human cancers.


Subject(s)
Cell Competition/genetics , Cell Competition/physiology , Drosophila/genetics , Drosophila/physiology , Animals , Cell Communication/genetics , Cell Communication/physiology , Epithelial Cells/physiology , Humans , Mutation/genetics , Oncogenes/genetics
15.
Dev Cell ; 53(4): 379-389.e5, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32386602

ABSTRACT

Metabolic diseases such as type 2 diabetes are associated with increased cancer incidence. Here, we show that hyperinsulinemia promotes epithelial tumorigenesis by abrogating cell competition. In Drosophila eye imaginal epithelium, oncogenic scribble (scrib) mutant cells are eliminated by cell competition when surrounded by wild-type cells. Through a genetic screen, we find that flies heterozygous for the insulin receptor substrate chico allow scrib cells to evade cell competition and develop into tumors. Intriguingly, chico is required in the brain's insulin-producing cells (IPCs) to execute cell competition remotely. Mechanistically, chico downregulation in IPCs causes hyperinsulinemia by upregulating a Drosophila insulin Dilp2, which activates insulin-mTOR signaling and thus boosts protein synthesis in scrib cells. A diet-induced increase in insulin levels also triggers scrib tumorigenesis, and pharmacological repression of protein synthesis prevents hyperinsulinemia-induced scrib overgrowth. Our findings provide an in vivo mechanistic link between metabolic disease and cancer risk via systemic regulation of cell competition.


Subject(s)
Carcinogenesis/pathology , Cell Competition , Diabetes Mellitus, Type 2/physiopathology , Drosophila melanogaster/metabolism , Hyperinsulinism/complications , Membrane Proteins/genetics , Neoplasms, Glandular and Epithelial/pathology , Tumor Suppressor Proteins/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Polarity , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Female , Humans , Male , Membrane Proteins/metabolism , Mutation , Neoplasms, Glandular and Epithelial/etiology , Neoplasms, Glandular and Epithelial/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism
16.
Sci Rep ; 9(1): 19549, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31863086

ABSTRACT

Multicellular organisms repair injured epithelium by evolutionarily conserved biological processes including activation of c-Jun N-terminal kinase (JNK) signaling. Here, we show in Drosophila imaginal epithelium that physical injury leads to the emergence of dying cells, which are extruded from the wounded tissue by JNK-induced Slit-Roundabout2 (Robo2) repulsive signaling. Reducing Slit-Robo2 signaling in the wounded tissue suppresses extrusion of dying cells and generates aberrant cells with highly upregulated growth factors Wingless (Wg) and Decapentaplegic (Dpp). The inappropriately elevated Wg and Dpp impairs wound repair, as halving one of these growth factor genes cancelled wound healing defects caused by Slit-Robo2 downregulation. Our data suggest that JNK-mediated Slit-Robo2 signaling contributes to epithelial wound repair by promoting extrusion of dying cells from the wounded tissue, which facilitates transient and appropriate induction of growth factors for proper wound healing.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/physiology , Animals , Drosophila/genetics , Drosophila Proteins/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Wound Healing/genetics , Wound Healing/physiology
17.
Dev Cell ; 51(1): 99-112.e4, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31543447

ABSTRACT

Cell competition is a quality control process that selectively eliminates unfit cells from the growing tissue via cell-cell interaction. Despite extensive mechanistic studies, the mechanism by which cell elimination is triggered has been elusive. Here, through a genetic screen in Drosophila, we discover that V-ATPase, an essential factor for autophagy, is required for triggering cell competition. Strikingly, autophagy is specifically elevated in prospective "loser" cells nearby wild-type "winner" cells, and blocking autophagy in loser cells abolishes their elimination. Mechanistically, elevated autophagy upregulates a proapoptotic gene hid through NFκB, and the elevated hid cooperates with JNK signaling to effectively induce loser's death. Crucially, this mechanism generally applies to cell competition caused by differences in protein synthesis between cells. Our findings establish a common mechanism of cell competition whereby cells with higher protein synthesis induce autophagy in their neighboring cells, leading to elimination of unfit cells.


Subject(s)
Autophagy , Drosophila melanogaster/genetics , MAP Kinase Kinase 4/metabolism , NF-kappa B/metabolism , Animals , Apoptosis , Binding, Competitive , Cell Communication , Cell Death , Cell Proliferation , Drosophila Proteins/metabolism , Female , Genotype , Male , Mutation , RNA Interference , Signal Transduction , Transcriptional Activation , Up-Regulation
18.
Dev Growth Differ ; 60(9): 522-530, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30443922

ABSTRACT

Cell competition is a context-dependent cell elimination through short-range cell-cell interaction, in which cells with higher fitness eliminate neighboring less-fit or oncogenic cells within the growing tissue. Cell competition can be triggered by many different factors such as heterozygous mutations in the ribosomal protein genes (which are called "Minute" mutations), elevated Myc, Yorkie/YAP, Wg/Wnt, JAK-STAT, Ras, or Src activity, and loss of Mahjong/VprBP, endocytic pathway components, or apicobasal cell polarity. Studies on the mechanisms and roles of cell competition have suggested that cell competition can be divided into two types: selection of fitter cells or elimination of oncogenic cells. The former type of cell competition includes Minute or Myc-induced cell competition that is considered to be dependent on the relative level of protein synthesis. The later type of cell competition includes tumor-suppressive cell competition triggered by loss of cell polarity genes such as scribble (scrib) or discs large (dlg). Genetic studies in Drosophila during the past decade have provided significant progress in understanding the mechanisms of these phenomena. At the same time, these studies have now raised new questions; how do different mechanisms contribute or cooperate to drive cell competition, do common mechanisms exist in different types of cell competition, and what are the physiological roles of these cell competition phenomena?


Subject(s)
Adaptation, Physiological/physiology , Cell Communication/physiology , Cell Proliferation/physiology , Models, Biological , Adaptation, Physiological/genetics , Animals , Cell Communication/genetics , Cell Proliferation/genetics , Humans , Mutation , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins pp60(c-src)/genetics , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction/genetics
19.
Curr Biol ; 28(13): 2115-2128.e5, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29910075

ABSTRACT

During the initial stage of tumor progression, oncogenic cells spread despite spatial confinement imposed by surrounding normal tissue. This spread of oncogenic cells (winners) is thought to be governed by selective killing of surrounding normal cells (losers) through a phenomenon called "cell competition" (i.e., supercompetition). Although the mechanisms underlying loser elimination are increasingly apparent, it is not clear how winner cells selectively occupy the space made available following loser apoptosis. Here, we combined live imaging analyses of two different oncogenic clones (Yki/YAP activation and Ras activation) in the Drosophila epithelium with computer simulation of tissue mechanics to elucidate such a mechanism. Contrary to the previous expectation that cell volume loss after apoptosis of loser cells was simply compensated for by the faster proliferation of winner cells, we found that the lost volume was compensated for by rapid cell expansion of winners. Mechanistically, the rapid winner-dominated cell expansion was driven by apoptosis-induced epithelial junction remodeling, which causes re-connection of local cellular connectivity (cell topology) in a manner that selectively increases winner apical surface area. In silico experiments further confirmed that repetition of loser elimination accelerates tissue-scale winner expansion through topological changes over time. Our proposed mechanism for linking loser death and winner expansion provides a new perspective on how tissue homeostasis disruption can initiate from an oncogenic mutation.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , Drosophila melanogaster/physiology , Epithelial Cells/physiology , Signal Transduction/physiology , Animals , Biomechanical Phenomena , Computer Simulation , Homeostasis
20.
Adv Exp Med Biol ; 1076: 173-194, 2018.
Article in English | MEDLINE | ID: mdl-29951820

ABSTRACT

Over the last few decades, Drosophila cancer models have made great contributions to our understanding toward fundamental cancer processes. Particularly, the development of genetic mosaic technique in Drosophila has enabled us to recapitulate basic aspects of human cancers, including clonal evolution, tumor microenvironment, cancer cachexia, and anticancer drug resistance. The mosaic technique has also led to the discovery of important tumor-suppressor pathways such as the Hippo pathway and the elucidation of the mechanisms underlying tumor growth and metastasis via regulation of cell polarity, cell-cell cooperation, and cell competition. Recent approaches toward identification of novel therapeutics using fly cancer models have further proved Drosophila as a robust system with great potentials for cancer research as well as anti-cancer therapy.


Subject(s)
Disease Models, Animal , Drosophila melanogaster , Neoplasms , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...