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










Publication year range
1.
bioRxiv ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38746415

ABSTRACT

Studies on Hippo pathway regulation of tumorigenesis largely center on YAP and TAZ, the transcriptional co-regulators of TEAD. Here, we present an oncogenic mechanism involving VGLL and TEAD fusions that is Hippo pathway-related but YAP/TAZ-independent. We characterize two recurrent fusions, VGLL2-NCOA2 and TEAD1-NCOA2, recently identified in spindle cell rhabdomyosarcoma. We demonstrate that, in contrast to VGLL2 and TEAD1, the fusion proteins are strong activators of TEAD-dependent transcription, and their function does not require YAP/TAZ. Furthermore, we identify that VGLL2 and TEAD1 fusions engage specific epigenetic regulation by recruiting histone acetyltransferase p300 to control TEAD-mediated transcriptional and epigenetic landscapes. We showed that small molecule p300 inhibition can suppress fusion proteins-induced oncogenic transformation both in vitro and in vivo. Overall, our study reveals a molecular basis for VGLL involvement in cancer and provides a framework for targeting tumors carrying VGLL, TEAD, or NCOA translocations.

2.
Sci Adv ; 9(21): eadc9660, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37224252

ABSTRACT

Adult stem cells are essential for tissue maintenance and repair. Although genetic pathways for controlling adult stem cells are extensively investigated in various tissues, much less is known about how mechanosensing could regulate adult stem cells and tissue growth. Here, we demonstrate that shear stress sensing regulates intestine stem cell proliferation and epithelial cell number in adult Drosophila. Ca2+ imaging in ex vivo midguts shows that shear stress, but not other mechanical forces, specifically activates enteroendocrine cells among all epithelial cell types. This activation is mediated by transient receptor potential A1 (TrpA1), a Ca2+-permeable channel expressed in enteroendocrine cells. Furthermore, specific disruption of shear stress, but not chemical, sensitivity of TrpA1 markedly reduces proliferation of intestinal stem cells and midgut cell number. Therefore, we propose that shear stress may act as a natural mechanical stimulation to activate TrpA1 in enteroendocrine cells, which, in turn, regulates intestine stem cell behavior.


Subject(s)
Adult Stem Cells , Drosophila Proteins , Drosophila , Ion Channels , Animals , Cell Proliferation , Intestines/cytology , Stress, Mechanical , Ion Channels/metabolism , Drosophila Proteins/metabolism
3.
Cell Rep ; 39(7): 110817, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35584678

ABSTRACT

Drosophila Toll-1 and all mammalian Toll-like receptors regulate innate immunity. However, the functions of the remaining eight Toll-related proteins in Drosophila are not fully understood. Here, we show that Drosophila Toll-9 is necessary and sufficient for a special form of compensatory proliferation after apoptotic cell loss (undead apoptosis-induced proliferation [AiP]). Mechanistically, for AiP, Toll-9 interacts with Toll-1 to activate the intracellular Toll-1 pathway for nuclear translocation of the NF-κB-like transcription factor Dorsal, which induces expression of the pro-apoptotic genes reaper and hid. This activity contributes to the feedback amplification loop that operates in undead cells. Given that Toll-9 also functions in loser cells during cell competition, we define a general role of Toll-9 in cellular stress situations leading to the expression of pro-apoptotic genes that trigger apoptosis and apoptosis-induced processes such as AiP. This work identifies conceptual similarities between cell competition and AiP.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Apoptosis/genetics , Cell Proliferation , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Feedback , Mammals/metabolism
4.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34620709

ABSTRACT

Germ cells possess the Piwi-interacting RNA pathway to repress transposable elements and maintain genome stability across generations. Transposable element mobilization in somatic cells does not affect future generations, but nonetheless can lead to pathological outcomes in host tissues. We show here that loss of function of the conserved zinc-finger transcription factor Hinfp causes dysregulation of many host genes and derepression of most transposable elements. There is also substantial DNA damage in somatic tissues of Drosophila after loss of Hinfp. Interference of transposable element mobilization by reverse-transcriptase inhibitors can suppress some of the DNA damage phenotypes. The key cell-autonomous target of Hinfp in this process is Histone1, which encodes linker histones essential for higher-order chromatin assembly. Transgenic expression of Hinfp or Histone1, but not Histone4 of core nucleosome, is sufficient to rescue the defects in repressing transposable elements and host genes. Loss of Hinfp enhances Ras-induced tissue growth and aging-related phenotypes. Therefore, Hinfp is a physiological regulator of Histone1-dependent silencing of most transposable elements, as well as many host genes, and serves as a venue for studying genome instability, cancer progression, neurodegeneration, and aging.


Subject(s)
DNA Transposable Elements/genetics , Drosophila melanogaster/genetics , Genomic Instability/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Developmental/genetics , Histones/genetics , Histones/metabolism , RNA, Small Interfering/genetics
5.
Stem Cell Reports ; 14(5): 828-844, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32330445

ABSTRACT

Tight junctions in mammals and septate junctions in insects are essential for epithelial integrity. We show here that, in the Drosophila intestine, smooth septate junction proteins provide barrier and signaling functions. During an RNAi screen for genes that regulate adult midgut tissue growth, we found that loss of two smooth septate junction components, Snakeskin and Mesh, caused a hyperproliferation phenotype. By examining epitope-tagged endogenous Snakeskin and Mesh, we demonstrate that the two proteins are present in the cytoplasm of differentiating enteroblasts and in cytoplasm and septate junctions of mature enterocytes. In both enteroblasts and enterocytes, loss of Snakeskin and Mesh causes Yorkie-dependent expression of the JAK-STAT pathway ligand Upd3, which in turn promotes proliferation of intestinal stem cells. Snakeskin and Mesh form a complex with each other, with other septate junction proteins and with Yorkie. Therefore, the Snakeskin-Mesh complex has both barrier and signaling function to maintain stem cell-mediated tissue homeostasis.


Subject(s)
Drosophila Proteins/metabolism , Enterocytes/metabolism , Gap Junctions/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Cell Differentiation , Cell Self Renewal , Cytoplasm/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster , Enterocytes/cytology , Homeostasis , Janus Kinases/metabolism , Membrane Proteins/genetics , Nuclear Proteins/genetics , STAT Transcription Factors/metabolism , Trans-Activators/genetics , YAP-Signaling Proteins
6.
Cell Stem Cell ; 26(5): 675-692.e8, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32259481

ABSTRACT

Intestinal homeostasis is tightly regulated by complex yet poorly understood signaling networks. Here, we demonstrate that Lats1/2, the core Hippo kinases, are essential to maintain Wnt pathway activity and intestinal stem cells. Lats1/2 deletion leads to loss of intestinal stem cells but drives Wnt-uncoupled crypt expansion. To explore the function of downstream transcriptional enhanced associate domain (TEAD) transcription factors, we identified a selective small-molecule reversible inhibitor of TEAD auto-palmitoylation that directly occupies its lipid-binding site and inhibits TEAD-mediated transcription in vivo. Combining this chemical tool with genetic and proteomics approaches, we show that intestinal Wnt inhibition by Lats deletion is Yes-associated protein (YAP)/transcriptional activator with PDZ-binding domain (TAZ) dependent but TEAD independent. Mechanistically, nuclear YAP/TAZ interact with Groucho/Transducin-Like Enhancer of Split (TLE) to block Wnt/T-cell factor (TCF)-mediated transcription, and dual inhibition of TEAD and Lats suppresses Wnt-uncoupled Myc upregulation and epithelial over-proliferation in Adenomatous polyposis coli (APC)-mutated intestine. Our studies highlight a pharmacological approach to inhibit TEAD palmitoylation and have important implications for targeting Wnt and Hippo signaling in human malignancies.


Subject(s)
Neoplasms , Transcription Factors , Humans , Intestines , Phosphoproteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Stem Cells/metabolism , Transcription Factors/metabolism
7.
Cell Rep ; 29(10): 3200-3211.e4, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31801083

ABSTRACT

Uveal melanoma (UM), the most common ocular malignancy, is characterized by GNAQ/11 mutations. Hippo/YAP and Ras/mitogen-activated protein kinase (MAPK) emerge as two important signaling pathways downstream of G protein alpha subunits of the Q class (GαQ/11)-mediated transformation, although whether and how they contribute to UM genesis in vivo remain unclear. Here, we adapt an adeno-associated virus (AAV)-based ocular injection method to directly deliver Cre recombinase into the mouse uveal tract and demonstrate that Lats1/2 kinases suppress UM formation specifically in uveal melanocytes. We find that genetic activation of YAP, but not Kras, is sufficient to initiate UM. We show that YAP/TAZ activation induced by Lats1/2 deletion cooperates with Kras to promote UM progression via downstream transcriptional reinforcement. Furthermore, dual inhibition of YAP/TAZ and Ras/MAPK synergizes to suppress oncogenic growth of human UM cells. Our data highlight the functional significance of Lats-YAP/TAZ in UM initiation and progression in vivo and suggest combination inhibition of YAP/TAZ and Ras/MAPK as a new therapeutic strategy for UM.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Cycle Proteins/genetics , Melanoma/genetics , Melanoma/pathology , Trans-Activators/genetics , Transcription Factors/genetics , Uveal Neoplasms/genetics , Uveal Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Female , HEK293 Cells , Humans , Melanocytes/pathology , Mice , Mitogen-Activated Protein Kinases/genetics , Mutation/genetics , Signal Transduction/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
8.
Genetics ; 212(4): 1227-1239, 2019 08.
Article in English | MEDLINE | ID: mdl-31213502

ABSTRACT

Colorectal cancer is a complex disease driven by well-established mutations such as APC and other yet to be identified pathways. The GTPase Rab11 regulates endosomal protein trafficking, and previously we showed that loss of Rab11 caused intestinal inflammation and hyperplasia in mice and flies. To test the idea that loss of Rab11 may promote cancer progression, we have analyzed archival human patient tissues and observed that 51 out of 70 colon cancer tissues had lower Rab11 protein staining. By using the Drosophila midgut model, we have found that loss of Rab11 can lead to three changes that may relate to cancer progression. First is the disruption of enterocyte polarity based on staining of the FERM domain protein Coracle. Second is an increased proliferation due to an increased expression of the JAK-STAT pathway ligand Upd3. Third is an increased expression of ImpL2, which is an IGFBP7 homolog and can suppress metabolism. Furthermore, loss of Rab11 can act synergistically with the oncoprotein RasV12 to regulate these cancer-related phenotypes.


Subject(s)
Colonic Neoplasms/genetics , Drosophila Proteins/genetics , rab GTP-Binding Proteins/genetics , Animals , Cell Polarity , Cell Proliferation , Colonic Neoplasms/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Enterocytes/cytology , Enterocytes/metabolism , Enterocytes/physiology , Humans , Insulin-Like Growth Factor Binding Proteins/genetics , Insulin-Like Growth Factor Binding Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , rab GTP-Binding Proteins/metabolism
9.
Cancer Res ; 79(16): 4099-4112, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31239271

ABSTRACT

The effects of polarized membrane trafficking in mature epithelial tissue on cell growth and cancer progression have not been fully explored in vivo. A majority of colorectal cancers have reduced and mislocalized Rab11, a small GTPase dedicated to trafficking of recycling endosomes. Patients with low Rab11 protein expression have poor survival rates. Using genetic models across species, we show that intact recycling endosome function restrains aberrant epithelial growth elicited by APC or RAS mutations. Loss of Rab11 protein led to epithelial dysplasia in early animal development and synergized with oncogenic pathways to accelerate tumor progression initiated by carcinogen, genetic mutation, or aging. Transcriptomic analysis uncovered an immediate expansion of the intestinal stem cell pool along with cell-autonomous Yki/Yap activation following disruption of Rab11a-mediated recycling endosomes. Intestinal tumors lacking Rab11a traffic exhibited marked elevation of nuclear Yap, upd3/IL6-Stat3, and amphiregulin-MAPK signaling, whereas suppression of Yki/Yap or upd3/IL6 reduced gut epithelial dysplasia and hyperplasia. Examination of Rab11a function in enteroids or cultured cell lines suggested that this endosome unit is required for suppression of the Yap pathway by Hippo kinases. Thus, recycling endosomes in mature epithelia constitute key tumor suppressors, loss of which accelerates carcinogenesis. SIGNIFICANCE: Recycling endosome traffic in mature epithelia constitutes a novel tumor suppressing mechanism.


Subject(s)
Colorectal Neoplasms/metabolism , Endosomes/metabolism , Epithelial Cells/pathology , rab GTP-Binding Proteins/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Animals, Genetically Modified , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Epithelial Cells/metabolism , Hippo Signaling Pathway , Humans , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Stem Cells/metabolism , Stem Cells/pathology , rab GTP-Binding Proteins/genetics
10.
J Biol Chem ; 294(26): 10172-10181, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31088910

ABSTRACT

The Toll signaling pathway in Drosophila melanogaster regulates several immune-related functions, including the expression of antimicrobial peptide (AMP) genes. The canonical Toll receptor (Toll-1) is activated by the cytokine Spätzle (Spz-1), but Drosophila encodes eight other Toll genes and five other Spz genes whose interactions with one another and associated functions are less well-understood. Here, we conducted in vitro assays in the Drosophila S2 cell line with the Toll/interleukin-1 receptor (TIR) homology domains of each Toll family member to determine whether they can activate a known target of Toll-1, the promoter of the antifungal peptide gene drosomycin. All TIR family members activated the drosomycin promoter, with Toll-1 and Toll-7 TIRs producing the highest activation. We found that the Toll-1 and Toll-7 ectodomains bind Spz-1, -2, and -5, and also vesicular stomatitis virus (VSV) virions, and that Spz-1, -2, -5, and VSV all activated the promoters of drosomycin and several other AMP genes in S2 cells expressing full-length Toll-1 or Toll-7. In vivo experiments indicated that Toll-1 and Toll-7 mutants could be systemically infected with two bacterial species (Enterococcus faecalis and Pseudomonas aeruginosa), the opportunistic fungal pathogen Candida albicans, and VSV with different survival times in adult females and males compared with WT fly survival. Our results suggest that all Toll family members can activate several AMP genes. Our results further indicate that Toll-1 and Toll-7 bind multiple Spz proteins and also VSV, but they differentially affect adult survival after systemic infection, potentially because of sex-specific differences in Toll-1 and Toll-7 expression.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Bacterial Infections/microbiology , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Gene Expression Regulation , Toll-Like Receptors/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Bacteria/isolation & purification , Bacterial Infections/genetics , Bacterial Infections/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/microbiology , Female , Male , Promoter Regions, Genetic , Signal Transduction , Toll-Like Receptors/genetics
11.
J Cell Physiol ; 234(12): 21925-21936, 2019 12.
Article in English | MEDLINE | ID: mdl-31042012

ABSTRACT

The intestinal epithelium has a high rate of cell turn over and is an excellent system to study stem cell-mediated tissue homeostasis. The Misshapen subfamily of the Ste20 kinases in mammals consists of misshapen like kinase 1 (MINK1), mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4), and TRAF2 and NCK interacting kinase (TNIK). Recent reports suggest that this subfamily has a novel function equal to the Hippo/MST subfamily as upstream kinases for Warts/Large tumor suppressor kinase (LATS) to suppress tissue growth. To study the in vivo functions of Mink1, Map4k4, and Tnik, we generated a compound knockout of these three genes in the mouse intestinal epithelium. The intestinal epithelia of the mutant animals were phenotypically normal up to approximately 12 months. The older animals then exhibited mildly increased proliferation throughout the lower GI tract. We also observed that the normally spatially organized Paneth cells in the crypt base became dispersed. The expression of one of the YAP pathway target genes Sox9 was increased while other target genes including CTGF did not show a significant change. Therefore, the Misshapen and Hippo subfamilies may have highly redundant functions to regulate growth in the intestinal epithelium, as illustrated in recent tissue culture models.


Subject(s)
Aging , Cell Proliferation/physiology , Intestinal Mucosa/metabolism , Stem Cells/metabolism , Animals , Mice, Transgenic , Phosphorylation/physiology
12.
Dev Cell ; 45(4): 433-449.e6, 2018 05 21.
Article in English | MEDLINE | ID: mdl-29754801

ABSTRACT

The intestinal epithelium has a high cell turnover rate and is an excellent system to study stem cell-mediated adaptive growth. In the Drosophila midgut, the Ste20 kinase Misshapen, which is distally related to Hippo, has a niche function to restrict intestinal stem cell activity. We show here that, under low growth conditions, Misshapen is localized near the cytoplasmic membrane, is phosphorylated at the threonine 194 by the upstream kinase Tao, and is more active toward Warts, which in turn inhibits Yorkie. Ingestion of yeast particles causes a midgut distention and a reduction of Misshapen membrane association and activity. Moreover, Misshapen phosphorylation is regulated by the stiffness of cell culture substrate, changing of actin cytoskeleton, and ingestion of inert particles. These results together suggest that dynamic membrane association and Tao phosphorylation of Misshapen are steps that link the mechanosensing of intestinal stretching after food particle ingestion to control adaptive growth.


Subject(s)
Cell Membrane/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Intestines/growth & development , Mechanotransduction, Cellular , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Trans-Activators/metabolism , Yeasts/metabolism , Adaptation, Physiological , Animals , Digestion , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Female , Intestinal Absorption , Intestinal Mucosa/metabolism , Male , Nuclear Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Trans-Activators/genetics , YAP-Signaling Proteins
13.
Dev Cell ; 43(1): 35-47.e4, 2017 10 09.
Article in English | MEDLINE | ID: mdl-28943241

ABSTRACT

YAP/TAZ are the major mediators of mammalian Hippo signaling; however, their precise function in the gastrointestinal tract remains poorly understood. Here we dissect the distinct roles of YAP/TAZ in endodermal epithelium and mesenchyme and find that, although dispensable for gastrointestinal epithelial development and homeostasis, YAP/TAZ function as the critical molecular switch to coordinate growth and patterning in gut mesenchyme. Our genetic analyses reveal that Lats1/2 kinases suppress expansion of the primitive mesenchymal progenitors, where YAP activation also prevents induction of the smooth muscle lineage through transcriptional repression of Myocardin. During later development, zone-restricted downregulation of YAP/TAZ provides the positional cue and allows smooth muscle cell differentiation induced by Hedgehog signaling. Taken together, our studies identify the mesenchymal requirement of YAP/TAZ in the gastrointestinal tract and highlight the functional interplays between Hippo and Hedgehog signaling underlying temporal and spatial control of tissue growth and specification in developing gut.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Differentiation/physiology , Epithelium/metabolism , Gastrointestinal Tract/metabolism , Hedgehog Proteins/metabolism , Mesoderm/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism , Acyltransferases , Animals , Cell Cycle Proteins , Epithelium/pathology , Mice, Transgenic , Nuclear Proteins/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , YAP-Signaling Proteins
14.
Dev Cell ; 36(3): 246-8, 2016 Feb 08.
Article in English | MEDLINE | ID: mdl-26859349

ABSTRACT

Toll and Hippo are two seemingly disparate pathways that regulate innate immunity and tissue growth, respectively. Reporting recently in Cell, Liu et al. (2016) tie them together by demonstrating that microbial infection activates Toll, which then signals to both pathways to modulate the antimicrobial response.


Subject(s)
Drosophila melanogaster/immunology , Immunity, Innate , Signal Transduction , Animals , Male
15.
Cell Stem Cell ; 17(6): 639-640, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26637937

ABSTRACT

The accumulation of somatic mutations in adult stem cells contributes to the decline of tissue functions and cancer initiation. In this issue of Cell Stem Cell, Siudeja et al. (2015) investigate the rate and mechanism of naturally occurring mutations in Drosophila midgut intestinal stem cells during aging and find high-frequency mutations arising from multiple mechanisms.


Subject(s)
Adult Stem Cells/metabolism , Drosophila/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intestinal Mucosa/metabolism , Mutation , Signal Transduction/genetics , Stem Cells/metabolism
16.
Stem Cell Rev Rep ; 11(6): 813-25, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26323255

ABSTRACT

The Drosophila adult midgut contains intestinal stem cells that support homeostasis and repair. We show here that the leucine zipper protein Bunched and the adaptor protein Madm are novel regulators of intestinal stem cells. MARCM mutant clonal analysis and cell type specific RNAi revealed that Bunched and Madm were required within intestinal stem cells for proliferation. Transgenic expression of a tagged Bunched showed a cytoplasmic localization in midgut precursors, and the addition of a nuclear localization signal to Bunched reduced its function to cooperate with Madm to increase intestinal stem cell proliferation. Furthermore, the elevated cell growth and 4EBP phosphorylation phenotypes induced by loss of Tuberous Sclerosis Complex or overexpression of Rheb were suppressed by the loss of Bunched or Madm. Therefore, while the mammalian homolog of Bunched, TSC-22, is able to regulate transcription and suppress cancer cell proliferation, our data suggest the model that Bunched and Madm functionally interact with the TOR pathway in the cytoplasm to regulate the growth and subsequent division of intestinal stem cells.


Subject(s)
Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Stem Cells/cytology , Tuberous Sclerosis/genetics , Tumor Suppressor Proteins/genetics , Animals , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/biosynthesis , Drosophila Proteins/metabolism , Intestines/cytology , Monomeric GTP-Binding Proteins/biosynthesis , Neuropeptides/biosynthesis , RNA Interference , RNA, Small Interfering/genetics , Ras Homolog Enriched in Brain Protein , Signal Transduction , Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis/metabolism , Tumor Suppressor Proteins/metabolism
17.
Cell Discov ; 1: 15038, 2015.
Article in English | MEDLINE | ID: mdl-27462435

ABSTRACT

The Hippo (Hpo) tumor suppressor pathway is an evolutionarily conserved signaling pathway that controls tissue growth and organ size in species ranging from Drosophila to human, and its malfunction has been implicated in many types of human cancer. In this study, we conducted a kinome screen and identified Happyhour (Hppy)/MAP4K3 as a novel player in the Hpo pathway. Our biochemical study showed that Hppy binds and phosphorylates Wts. Our genetic experiments suggest that Hppy acts in parallel and partial redundantly with Misshapen (Msn)/MAP4K4 to regulate Yki nuclear localization and Hpo target gene expression in Drosophila wing imaginal discs. Furthermore, we showed that cytoskeleton stress restricts Yki nuclear localization through Hppy and Msn when Hpo activity is compromised, thus providing an explanation for the Wts-dependent but Hpo-independent regulation of Yki in certain contexts. Our study has unraveled an additional layer of complexity in the Hpo signaling pathway and laid down a foundation for exploring how different upstream regulators feed into the core Hpo pathway.

18.
Dev Cell ; 31(3): 291-304, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25453828

ABSTRACT

Similar to the mammalian intestine, the Drosophila adult midgut has resident stem cells that support growth and regeneration. How the niche regulates intestinal stem cell activity in both mammals and flies is not well understood. Here, we show that the conserved germinal center protein kinase Misshapen restricts intestinal stem cell division by repressing the expression of the JAK-STAT pathway ligand Upd3 in differentiating enteroblasts. Misshapen, a distant relative to the prototypic Warts activating kinase Hippo, interacts with and activates Warts to negatively regulate the activity of Yorkie and the expression of Upd3. The mammalian Misshapen homolog MAP4K4 similarly interacts with LATS (Warts homolog) and promotes inhibition of YAP (Yorkie homolog). Together, this work reveals that the Misshapen-Warts-Yorkie pathway acts in enteroblasts to control niche signaling to intestinal stem cells. These findings also provide a model in which to study requirements for MAP4K4-related kinases in MST1/2-independent regulation of LATS and YAP.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intestinal Mucosa/metabolism , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Regeneration/physiology , Signal Transduction/physiology , Trans-Activators/metabolism , Animals , Cell Differentiation/physiology , Cell Division , Regeneration/genetics , Stem Cells/cytology , YAP-Signaling Proteins
19.
Cell Rep ; 9(1): 32-39, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25263551

ABSTRACT

Intestinal stem cells in the adult Drosophila midgut are regulated by growth factors produced from the surrounding niche cells including enterocytes and visceral muscle. The role of the other major cell type, the secretory enteroendocrine cells, in regulating intestinal stem cells remains unclear. We show here that newly eclosed scute loss-of-function mutant flies are completely devoid of enteroendocrine cells. These enteroendocrine cell-less flies have normal ingestion and fecundity but shorter lifespan. Moreover, in these newly eclosed mutant flies, the diet-stimulated midgut growth that depends on the insulin-like peptide 3 expression in the surrounding muscle is defective. The depletion of Tachykinin-producing enteroendocrine cells or knockdown of Tachykinin leads to a similar although less severe phenotype. These results establish that enteroendocrine cells serve as an important link between diet and visceral muscle expression of an insulin-like growth factor to stimulate intestinal stem cell proliferation and tissue growth.


Subject(s)
Enteroendocrine Cells/physiology , Stem Cells/physiology , Animals , Cell Differentiation/physiology , Drosophila , Enterocytes/metabolism , Enterocytes/physiology , Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Female , Homeostasis , Intestinal Mucosa/metabolism , Intestines/cytology , Male , Stem Cells/cytology , Stem Cells/metabolism , Tachykinins/metabolism
20.
Development ; 141(13): 2621-32, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24961799

ABSTRACT

Intestinal stem cells (ISCs) in the adult Drosophila midgut can respond to tissue damage and support repair. We used genetic manipulation to increase the number of ISC-like cells in the adult midgut and performed gene expression profiling to identify potential ISC regulators. A detailed analysis of one of these potential regulators, the zinc-finger protein Charlatan, was carried out. MARCM clonal analysis and RNAi in precursor cells showed that loss of Chn function caused severe ISC division defects, including loss of EdU incorporation, phosphorylated histone 3 staining and expression of the mitotic protein Cdc2. Loss of Charlatan also led to a much reduced histone acetylation staining in precursor cells. Both the histone acetylation and ISC division defects could be rescued by the simultaneous decrease of the Histone Deacetylase 2. The overexpression of Charlatan blocked differentiation reversibly, but loss of Charlatan did not lead to automatic differentiation. The results together suggest that Charlatan does not simply act as an anti-differentiation factor but instead functions to maintain a chromatin structure that is compatible with stem cell properties, including proliferation.


Subject(s)
Cell Differentiation/physiology , Drosophila Proteins/physiology , Drosophila/genetics , Intestines/cytology , Stem Cells/physiology , Transcription Factors/physiology , Animals , Cell Differentiation/genetics , Drosophila/physiology , Gene Expression Profiling , Microarray Analysis , Microscopy, Fluorescence , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL