Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Sci Signal ; 16(807): eadd6834, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37847757

ABSTRACT

Hedgehog (Hh) signaling controls growth and patterning during embryonic development and homeostasis in adult tissues. Hh binding to the receptor Patched (Ptc) elicits intracellular signaling by relieving Ptc-mediated inhibition of the transmembrane protein Smoothened (Smo). We uncovered a role for the lipid phosphatidic acid (PA) in the regulation of the Hh pathway in Drosophila melanogaster. Deleting the Ptc C-terminal tail or mutating the predicted PA-binding sites within it prevented Ptc from inhibiting Smo in wing discs and in cultured cells. The C-terminal tail of Ptc directly interacted with PA in vitro, an association that was reduced by Hh, and increased the amount of PA at the plasma membrane in cultured cells. Smo also interacted with PA in vitro through a binding pocket located in the transmembrane region, and mutating residues in this pocket reduced Smo activity in vivo and in cells. By genetically manipulating PA amounts in vivo or treating cultured cells with PA, we demonstrated that PA promoted Smo activation. Our findings suggest that Ptc may sequester PA in the absence of Hh and release it in the presence of Hh, thereby increasing the amount of PA that is locally available to promote Smo activation.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Patched Receptors/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor/genetics , Smoothened Receptor/metabolism
2.
Front Cell Dev Biol ; 10: 847844, 2022.
Article in English | MEDLINE | ID: mdl-35321245

ABSTRACT

The GPCR-family protein Smoothened (Smo) is essential for Hedgehog (Hh) signal transduction in both insects and vertebrates. The regulation of subcellular localization and abundance of Smo is a critical step in Hh signaling. Recent studies have demonstrated that Smo is subjected to ubiquitination mediated by multiple E3 ubiquitin ligases, leading to Smo endocytosis and subsequent degradation through the proteasome- and lysosome-mediated pathways in Drosophila. Ubiquitination of Smo also promotes its ciliary exit in mammalian cells. Hh inhibits Smo ubiquitination by blocking E3 ligase recruitment and promoting Smo deubiquitination through the ubiquitin-specific protease 8 (USP8) in Drosophila. Inhibition of Smo ubiquitination by Hh promotes Smo cell surface accumulation in Drosophila and ciliary accumulation in mammalian cells. Interestingly, Hh also induces sumoylation of Smo in both Drosophila and mammalian cells, which promotes Smo cell surface/ciliary accumulation. This review focuses on how ubiquitination and sumoylation regulate Smo intracellular trafficking and abundance and how these processes are regulated by Hh.

3.
Cell Mol Gastroenterol Hepatol ; 13(2): 501-516, 2022.
Article in English | MEDLINE | ID: mdl-34560309

ABSTRACT

BACKGROUND & AIMS: Intestinal stem cells (ISCs) are sensitive to dietary alterations and nutrient availability. Neurotensin (NT), a gut peptide localized predominantly to the small bowel and released by fat ingestion, stimulates the growth of intestinal mucosa under basal conditions and during periods of nutrient deprivation, suggesting a possible role for NT on ISC function. METHODS: Leucine-rich repeat-containing G-protein coupled receptor 5-Enhanced Green Fluorescent Protein (Lgr5-EGFP) NT wild type (Nt+/+) and Lgr5-EGFP NT knockout (Nt-/-) mice were fed ad libitum or fasted for 48 hours. Small intestine tissue and crypts were examined by gene expression analyses, fluorescence-activated cell sorting, Western blot, immunohistochemistry, and crypt-derived organoid culture. Drosophila expressing NT in midgut enteroendocrine cells were fed a standard diet or low-energy diet and esg-green fluorescent protein+ ISCs were quantified via immunofluorescence. RESULTS: Loss of NT impaired crypt cell proliferation and ISC function in a manner dependent on nutrient status. Under nutrient-rich conditions, NT stimulated extracellular signal-regulated kinases 1 and 2 signaling and the expression of genes that promote cell-cycle progression, leading to crypt cell proliferation. Under conditions of nutrient depletion, NT stimulated WNT/ß-catenin signaling and promoted an ISC gene signature, leading to enhanced ISC function. NT was required for the induction of WNT/ß-catenin signaling and ISC-specific gene expression during nutrient depletion, and loss of NT reduced crypt cell proliferation and impaired ISC function and Lgr5 expression in the intestine during fasting. Conversely, the expression of NT in midgut enteroendocrine cells of Drosophila prevented loss of ISCs during nutrient depletion. CONCLUSIONS: Collectively, our findings establish an evolutionarily conserved role for NT in ISC maintenance during nutritional stress. GSE182828.


Subject(s)
Neurotensin , Stem Cells , Animals , Cell Proliferation , Intestine, Small , Mice , Neurotensin/metabolism , Nutrients , Stem Cells/metabolism
4.
Methods Mol Biol ; 2374: 121-137, 2022.
Article in English | MEDLINE | ID: mdl-34562248

ABSTRACT

The GPCR-family protein Smoothened (Smo) is an obligatory signal transducer of the Hedgehog (Hh) signaling pathway. Binding of Hh to its receptor Patched (Ptc) alleviates Ptc-mediated inhibition of Smo, allowing Smo to activate the Cubitus interruptus (Ci)/Gli family of zinc finger transcription factors. The activation of Smo is an early and crucial event in Hh signal transduction. Studies have shown that Hh induces cell surface/ciliary accumulation and phosphorylation of Smo by multiple kinases, including protein kinase A (PKA), casein kinase 1 (CK1), casein kinase 2 (CK2), G protein-coupled receptor kinase 2 (Gprk2/GRK2), and atypical PKC (aPKC). Here, we describe the assays used to examine the phosphorylation and activity of Smo, including in vitro kinase assay, phospho-specific antibodies, luciferase reporter assay, cell surface accumulation, and ciliary localization assays. These assays provide powerful tools to study Smo phosphorylation and activation, leading to mechanistic insight into Smo regulation.


Subject(s)
Smoothened Receptor/metabolism , Drosophila Proteins/metabolism , Hedgehog Proteins/metabolism , Phosphorylation , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Smoothened Receptor/genetics , Transcription Factors/metabolism
5.
Cells ; 10(8)2021 08 20.
Article in English | MEDLINE | ID: mdl-34440907

ABSTRACT

The seven-transmembrane protein, Smoothened (SMO), has shown to be critical for the hedgehog (HH) signal transduction on the cell membrane (and the cilium in vertebrates). SMO is subjected to multiple types of post-translational regulations, including phosphorylation, ubiquitination, and sumoylation, which alter SMO intracellular trafficking and cell surface accumulation. Recently, SMO is also shown to be regulated by small molecules, such as oxysterol, cholesterol, and phospholipid. The activity of SMO must be very well balanced by these different mechanisms in vivo because the malfunction of SMO will not only cause developmental defects in early stages, but also induce cancers in late stages. Here, we discuss the activation and inactivation of SMO by different mechanisms to better understand how SMO is regulated by the graded HH signaling activity that eventually governs distinct development outcomes.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Hedgehog Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Smoothened Receptor/metabolism , Animals , Cell Membrane/metabolism , Cilia/metabolism , Models, Biological , Receptors, Cell Surface/metabolism
6.
FASEB J ; 34(6): 8596-8610, 2020 06.
Article in English | MEDLINE | ID: mdl-32359121

ABSTRACT

We previously reported that high levels of plasma neurotensin (NT), a gut hormone released from enteroendocrine cells of the small bowel, contribute to obesity and comorbid conditions. Gut microbiota has been implicated in the obesity development. Paneth cells are critical in maintaining gut microbiota composition and homeostasis by releasing antimicrobial proteins including α-defensins. The purpose of our current study was to determine the possible role of NT in gut microbiota composition and α-defensin gene expression associated with obesity. Here we show that the ratio of Firmicutes/Bacteroidetes (F/B ratio) and intestinal proinflammatory cytokines is significantly increased in NT+/+ mice fed with a high-fat diet (HFD) which were improved in NT-deficient mice. HFD disrupted the intestinal Mmp7/α-defensin axis, which was completely prevented in NT-/- mice. In addition, NT treatment inhibited DEFA5 expression and concurrent NF-κB activity, which was blocked by a pan PKC inhibitor (Gö6983) or an inhibitor for atypical PKCs (CRT0066854). More importantly, the shRNA-mediated knockdown of atypical PKCτ reversed NT-attenuated DEFA5 expression and increased NF-κB activity. NT contributes to the HFD-induced disruption of gut microbiota composition and α-defensin expression. PKCτ/λ plays a central role in NT-mediated α-defensin gene expression which might be mediated through the inhibition of NF-κB signaling pathways in Paneth cells.


Subject(s)
Dysbiosis/metabolism , Inflammation/metabolism , Matrix Metalloproteinase 7/metabolism , Neurotensin/metabolism , alpha-Defensins/metabolism , Adipose Tissue/metabolism , Animals , Cytokines/metabolism , Diet, High-Fat/adverse effects , Dysbiosis/pathology , Gastrointestinal Microbiome/physiology , Inflammation/pathology , Insulin Resistance/physiology , Intestines/pathology , Male , Mice , Mice, Obese , NF-kappa B/metabolism , Obesity/metabolism , Paneth Cells/metabolism , Signal Transduction/physiology
7.
Dev Biol ; 457(1): 128-139, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31550483

ABSTRACT

Hedgehog (Hh) signaling has been shown to regulate multiple developmental processes, however, it is unclear how it regulates lipid metabolism. Here, we demonstrate that Hh signaling exhibits potent activity in Drosophila fat body, which is induced by both locally expressed and midgut-derived Hh proteins. Inactivation of Hh signaling increases, whereas activation of Hh signaling decreases lipid accumulation. The major lipase Brummer (Bmm) acts downstream of Smoothened (Smo) in Hh signaling to promote lipolysis, therefore, the breakdown of triacylglycerol (TAG). We identify a critical Ci binding site in bmm promoter that is responsible to mediate Bmm expression induced by Hh signaling. Genomic mutation of the Ci binding site significantly reduces the expression of Bmm and dramatically decreases the responsiveness to Ci overexpression. Together, our findings provide a model for lipolysis to be regulated by Hh signaling, raising the possibility for Hh signaling to be involved in lipid metabolic and/or lipid storage diseases.


Subject(s)
Drosophila Proteins/genetics , Drosophila/metabolism , Lipase/genetics , Lipolysis , Signal Transduction , Adipocytes/metabolism , Animals , DNA-Binding Proteins/metabolism , Drosophila/growth & development , Drosophila Proteins/metabolism , Fat Body/metabolism , Female , Hedgehog Proteins/metabolism , Larva/metabolism , Male , Smoothened Receptor/metabolism , Transcription Factors/metabolism
8.
J Cell Sci ; 131(1)2018 01 04.
Article in English | MEDLINE | ID: mdl-29142103

ABSTRACT

Smoothened (Smo), a GPCR family protein, plays a critical role in the reception and transduction of Hedgehog (Hh) signal. Smo is phosphorylated and activated on the cell surface; however, it is unknown whether Smo can be intracellularly activated. Here, we demonstrate that inactivation of the ESCRT-III causes dramatic accumulation of Smo in the ESCRT-III/MVB compartment, and subsequent activation of Hh signaling. In contrast, inactivation of ESCRTs 0-II induces mild Smo accumulation in the ESCRT-III/MVB compartment. We provide evidence that Kurtz (Krz), the Drosophila ß-arrestin2, acts in parallel with the ESCRTs 0-II pathway to sort Smo to the multivesicular bodies and lysosome-mediated degradation. Additionally, upon inactivation of ESCRT-III, all active and inactive forms of Smo are accumulated. Endogenous Smo accumulated upon ESCRT-III inactivation is highly activated, which is induced by phosphorylation but not sumoylation. Taken together, our findings demonstrate a model for intracellular activation of Smo, raising the possibility for tissue overgrowth caused by an excessive amount, rather than mutation of Smo.


Subject(s)
Drosophila Proteins/metabolism , Drosophila/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Smoothened Receptor/metabolism , Animals , Arrestins/genetics , Arrestins/metabolism , Cell Membrane/metabolism , Drosophila/cytology , Drosophila/growth & development , Drosophila Proteins/genetics , Endosomal Sorting Complexes Required for Transport/genetics , Phosphorylation , Protein Transport/genetics , Smoothened Receptor/genetics , Sumoylation
9.
Stem Cell Reports ; 8(4): 991-1004, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28330618

ABSTRACT

Natural genetic diversity offers an important yet largely untapped resource to decipher the molecular mechanisms regulating hematopoietic stem cell (HSC) function. Latexin (Lxn) is a negative stem cell regulatory gene identified on the basis of genetic diversity. By using an Lxn knockout mouse model, we found that Lxn inactivation in vivo led to the physiological expansion of the entire hematopoietic hierarchy. Loss of Lxn enhanced the competitive repopulation capacity and survival of HSCs in a cell-intrinsic manner. Gene profiling of Lxn-null HSCs showed altered expression of genes enriched in cell-matrix and cell-cell interactions. Thrombospondin 1 (Thbs1) was a potential downstream target with a dramatic downregulation in Lxn-null HSCs. Enforced expression of Thbs1 restored the Lxn inactivation-mediated HSC phenotypes. This study reveals that Lxn plays an important role in the maintenance of homeostatic hematopoiesis, and it may lead to development of safe and effective approaches to manipulate HSCs for clinical benefit.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Nerve Tissue Proteins/genetics , Animals , Cell Proliferation , Cell Survival , Gene Deletion , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Sci Rep ; 7: 42749, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28195188

ABSTRACT

In Hedgehog (Hh) signaling, the GPCR-family protein Smoothened (Smo) acts as a signal transducer that is regulated by phosphorylation and ubiquitination, which ultimately change the cell surface accumulation of Smo. However, it is not clear whether Smo is regulated by other post-translational modifications, such as sumoylation. Here, we demonstrate that knockdown of the small ubiquitin-related modifier (SUMO) pathway components Ubc9 (a SUMO-conjugating enzyme E2), PIAS (a SUMO-protein ligase E3), and Smt3 (the SUMO isoform in Drosophila) by RNAi prevents Smo accumulation and alters Smo activity in the wing. We further show that Hh-induced-sumoylation stabilizes Smo, whereas desumoylation by Ulp1 destabilizes Smo in a phosphorylation independent manner. Mechanistically, we discover that excessive Krz, the Drosophila ß-arrestin 2, inhibits Smo sumoylation and prevents Smo accumulation through Krz regulatory domain. Krz likely facilitates the interaction between Smo and Ulp1 because knockdown of Krz by RNAi attenuates Smo-Ulp1 interaction. Finally, we provide evidence that Cos2 is also sumoylated, which counteracts its inhibitory role on Smo accumulation in the wing. Taken together, we have uncovered a novel mechanism for Smo activation by sumoylation that is regulated by Hh and Smo interacting proteins.


Subject(s)
Drosophila Proteins/metabolism , Kinesins/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Smoothened Receptor/metabolism , Sumoylation , Animals , Arrestins/genetics , Arrestins/metabolism , Cell Line , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Drosophila , Drosophila Proteins/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Kinesins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/genetics , Smoothened Receptor/genetics , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Wings, Animal/metabolism
11.
Nat Commun ; 8: 14228, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28198361

ABSTRACT

Snail1, a key transcription factor of epithelial-mesenchymal transition (EMT), is subjected to ubiquitination and degradation, but the mechanism by which Snail1 is stabilized in tumours remains unclear. We identify Dub3 as a bona fide Snail1 deubiquitinase, which interacts with and stabilizes Snail1. Dub3 is overexpressed in breast cancer; knockdown of Dub3 resulted in Snail1 destabilization, suppressed EMT and decreased tumour cell migration, invasion, and metastasis. These effects are rescued by ectopic Snail1 expression. IL-6 also stabilizes Snail1 by inducing Dub3 expression, the specific inhibitor WP1130 binds to Dub3 and inhibits the Dub3-mediating Snail1 stabilization in vitro and in vivo. Our study reveals a critical Dub3-Snail1 signalling axis in EMT and metastasis, and provides an effective therapeutic approach against breast cancer.


Subject(s)
Breast Neoplasms/pathology , Endopeptidases/metabolism , Proteolysis , Snail Family Transcription Factors/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Down-Regulation/genetics , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Interleukin-6/metabolism , Mice, SCID , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Binding , Protein Stability , Protein Transport , Ubiquitination
12.
Nature ; 533(7603): 411-5, 2016 05 19.
Article in English | MEDLINE | ID: mdl-27193687

ABSTRACT

Obesity and its associated comorbidities (for example, diabetes mellitus and hepatic steatosis) contribute to approximately 2.5 million deaths annually and are among the most prevalent and challenging conditions confronting the medical profession. Neurotensin (NT; also known as NTS), a 13-amino-acid peptide predominantly localized in specialized enteroendocrine cells of the small intestine and released by fat ingestion, facilitates fatty acid translocation in rat intestine, and stimulates the growth of various cancers. The effects of NT are mediated through three known NT receptors (NTR1, 2 and 3; also known as NTSR1, 2, and NTSR3, respectively). Increased fasting plasma levels of pro-NT (a stable NT precursor fragment produced in equimolar amounts relative to NT) are associated with increased risk of diabetes, cardiovascular disease and mortality; however, a role for NT as a causative factor in these diseases is unknown. Here we show that NT-deficient mice demonstrate significantly reduced intestinal fat absorption and are protected from obesity, hepatic steatosis and insulin resistance associated with high fat consumption. We further demonstrate that NT attenuates the activation of AMP-activated protein kinase (AMPK) and stimulates fatty acid absorption in mice and in cultured intestinal cells, and that this occurs through a mechanism involving NTR1 and NTR3 (also known as sortilin). Consistent with the findings in mice, expression of NT in Drosophila midgut enteroendocrine cells results in increased lipid accumulation in the midgut, fat body, and oenocytes (specialized hepatocyte-like cells) and decreased AMPK activation. Remarkably, in humans, we show that both obese and insulin-resistant subjects have elevated plasma concentrations of pro-NT, and in longitudinal studies among non-obese subjects, high levels of pro-NT denote a doubling of the risk of developing obesity later in life. Our findings directly link NT with increased fat absorption and obesity and suggest that NT may provide a prognostic marker of future obesity and a potential target for prevention and treatment.


Subject(s)
Diet, High-Fat/adverse effects , Neurotensin/metabolism , Obesity/chemically induced , Obesity/metabolism , AMP-Activated Protein Kinases/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Disease Models, Animal , Drosophila melanogaster/cytology , Drosophila melanogaster/enzymology , Drosophila melanogaster/metabolism , Enteroendocrine Cells/metabolism , Enzyme Activation , Fat Body/metabolism , Fatty Acids/metabolism , Fatty Liver/metabolism , Fatty Liver/prevention & control , Female , Humans , Insulin Resistance/physiology , Intestinal Mucosa/metabolism , Intestines/cytology , Lipid Metabolism , Male , Mice , Middle Aged , Neurotensin/blood , Neurotensin/deficiency , Neurotensin/genetics , Obesity/blood , Obesity/prevention & control , Protein Precursors/blood , Protein Precursors/metabolism
13.
PLoS Genet ; 12(5): e1006054, 2016 05.
Article in English | MEDLINE | ID: mdl-27195754

ABSTRACT

Hedgehog (Hh) signaling regulates multiple aspects of metazoan development and tissue homeostasis, and is constitutively active in numerous cancers. We identified Ubr3, an E3 ubiquitin ligase, as a novel, positive regulator of Hh signaling in Drosophila and vertebrates. Hh signaling regulates the Ubr3-mediated poly-ubiquitination and degradation of Cos2, a central component of Hh signaling. In developing Drosophila eye discs, loss of ubr3 leads to a delayed differentiation of photoreceptors and a reduction in Hh signaling. In zebrafish, loss of Ubr3 causes a decrease in Shh signaling in the developing eyes, somites, and sensory neurons. However, not all tissues that require Hh signaling are affected in zebrafish. Mouse UBR3 poly-ubiquitinates Kif7, the mammalian homologue of Cos2. Finally, loss of UBR3 up-regulates Kif7 protein levels and decreases Hh signaling in cultured cells. In summary, our work identifies Ubr3 as a novel, evolutionarily conserved modulator of Hh signaling that boosts Hh in some tissues.


Subject(s)
Drosophila Proteins/genetics , Eye/metabolism , Kinesins/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Eye/growth & development , Hedgehog Proteins/genetics , Kinesins/metabolism , Mice , Photoreceptor Cells/metabolism , Polyubiquitin , Proteolysis , RNA, Small Interfering , Signal Transduction , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Zebrafish/genetics
14.
PLoS Biol ; 14(2): e1002375, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26863604

ABSTRACT

In Hedgehog (Hh) signaling, binding of Hh to the Patched-Interference Hh (Ptc-Ihog) receptor complex relieves Ptc inhibition on Smoothened (Smo). A longstanding question is how Ptc inhibits Smo and how such inhibition is relieved by Hh stimulation. In this study, we found that Hh elevates production of phosphatidylinositol 4-phosphate (PI(4)P). Increased levels of PI(4)P promote, whereas decreased levels of PI(4)P inhibit, Hh signaling activity. We further found that PI(4)P directly binds Smo through an arginine motif, which then triggers Smo phosphorylation and activation. Moreover, we identified the pleckstrin homology (PH) domain of G protein-coupled receptor kinase 2 (Gprk2) as an essential component for enriching PI(4)P and facilitating Smo activation. PI(4)P also binds mouse Smo (mSmo) and promotes its phosphorylation and ciliary accumulation. Finally, Hh treatment increases the interaction between Smo and PI(4)P but decreases the interaction between Ptc and PI(4)P, indicating that, in addition to promoting PI(4)P production, Hh regulates the pool of PI(4)P associated with Ptc and Smo.


Subject(s)
Drosophila Proteins/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , Hedgehog Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cilia/metabolism , Drosophila , Mice , NIH 3T3 Cells , Patched Receptors , Patched-1 Receptor , Phosphorylation , Receptors, Cell Surface/metabolism , Smoothened Receptor
15.
Methods Mol Biol ; 1322: 45-60, 2015.
Article in English | MEDLINE | ID: mdl-26179038

ABSTRACT

Smoothened (Smo) is essential for transduction of the Hedgehog (Hh) signal in both insects and vertebrates. Binding of Hh to Ptc-Ihog relieves the Patched (Ptc)-mediated inhibition of Smo, which allows Smo to activate the cubitus interruptus (Ci)/Gli family of zinc finger transcription factors and thereby induce the expression of Hh target genes, such as decapentaplegic (dpp), ptc, and engrailed (en). The activation of Smo appears to be one of the most important events in Hh signaling. Studies have shown that Hh induces cell surface/ciliary accumulation and phosphorylation of Smo by multiple kinases, including protein kinase A (PKA), casein kinase 1 (CK1), casein kinase 2 (CK2), G protein-coupled receptor kinase 2 (Gprk2), and atypical PKC (aPKC). Here, we describe the assays used to examine the activity of Smo in Hh signaling, including in vitro kinase, ptc-luciferase reporter assay, cell surface accumulation assay, fluorescence resonance energy transfer (FRET) assay, and wing disc immunostaining. These assays are powerful tools to study Smo phosphorylation and activation, which have provided mechanistic insight into a better understanding the mechanisms of Smo regulation.


Subject(s)
Drosophila Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Wings, Animal/metabolism , Animals , Animals, Genetically Modified , Cell Membrane/metabolism , Cells, Cultured , Drosophila , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Gene Expression , Gene Targeting , Genes, Reporter , Mutation , Phosphorylation , Protein Interaction Domains and Motifs , Protein Multimerization , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins , Smoothened Receptor , Wings, Animal/growth & development
16.
Mol Cell Biol ; 35(19): 3301-11, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26169834

ABSTRACT

Wnt signaling plays important roles in development and tumorigenesis. A central question about the Wnt pathway is the regulation of ß-catenin. Phosphorylation of ß-catenin by CK1α and GSK3 promotes ß-catenin binding to ß-TrCP, leading to ß-catenin degradation through the proteasome. The phosphorylation and ubiquitination of ß-catenin have been well characterized; however, it is unknown whether and how a deubiquitinase is involved. In this study, by screening RNA interference (RNAi) libraries, we identified USP47 as a deubiquitinase that prevents ß-catenin ubiquitination. Inactivation of USP47 by RNAi increased ß-catenin ubiquitination, attenuated Wnt signaling, and repressed cancer cell growth. Furthermore, USP47 deubiquitinates itself, whereas ß-TrCP promotes USP47 ubiquitination through interaction with an atypical motif in USP47. Finally, in vivo studies in the Drosophila wing suggest that UBP64E, the USP47 counterpart in Drosophila, is required for Armadillo stabilization and plays a positive role in regulating Wnt target gene expression.


Subject(s)
Drosophila Proteins/physiology , Ubiquitin Thiolesterase/physiology , Ubiquitin-Specific Proteases/physiology , Ubiquitination , Wnt Signaling Pathway , beta Catenin/metabolism , Amino Acid Sequence , Animals , Binding Sites , Drosophila melanogaster , HEK293 Cells , Humans , Molecular Sequence Data , Proteolysis , Wings, Animal/enzymology , beta-Transducin Repeat-Containing Proteins/metabolism
17.
PLoS Pathog ; 11(3): e1004783, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25815785

ABSTRACT

Stimulator of interferon genes (STING, also known as MITA and ERIS) is critical in protecting the host against DNA pathogen invasion. However, the molecular mechanism underlying the regulation of STING remains unclear. Here, we show that PPM1A negatively regulates antiviral signaling by targeting STING in its phosphatase activity-dependent manner, and in a line with this, PPM1A catalytically dephosphorylates STING and TBK1 in vitro. Importantly, we provide evidence that whereas TBK1 promotes STING aggregation in a phosphorylation-dependent manner, PPM1A antagonizes STING aggregation by dephosphorylating both STING and TBK1, emphasizing that phosphorylation is crucial for the efficient activation of STING. Our findings demonstrate a novel regulatory circuit in which STING and TBK1 reciprocally regulate each other to enable efficient antiviral signaling activation, and PPM1A dephosphorylates STING and TBK1, thereby balancing this antiviral signal transduction.


Subject(s)
Membrane Proteins/immunology , Phosphoprotein Phosphatases/immunology , Protein Serine-Threonine Kinases/immunology , Signal Transduction/immunology , Animals , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Phosphorylation/genetics , Phosphorylation/immunology , Protein Phosphatase 2C , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , Vero Cells
18.
Front Biol (Beijing) ; 10(6): 475-486, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26973699

ABSTRACT

The Hedgehog (Hh) signaling pathway play critical roles in embryonic development and adult tissue homeostasis. A critical step in Hh signal transduction is how Hh receptor Patched (Ptc) inhibits the atypical G protein-coupled receptor Smoothened (Smo) in the absence of Hh and how this inhibition is release by Hh stimulation. It is unlikely that Ptc inhibits Smo by direct interaction. Here we discuss how Hh regulates the phosphorylation and ubiquitination of Smo, leading to cell surface and ciliary accumulation of Smo in Drosophila and vertebrate cells, respectively. In addition, we discuss how PI(4)P phospholipid acts in between Ptc and Smo to regulate Smo phosphorylation and activation in response to Hh stimulation.

19.
Proc Natl Acad Sci U S A ; 111(45): E4842-50, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25349414

ABSTRACT

Smoothened (Smo) is essential for transduction of the Hedgehog (Hh) signal in both insects and vertebrates. Cell surface/cilium accumulation of Smo is thought to play an important role in Hh signaling, but how the localization of Smo is controlled remains poorly understood. In this study, we demonstrate that atypical PKC (aPKC) regulates Smo phosphorylation and basolateral accumulation in Drosophila wings. Inactivation of aPKC by either RNAi or a mutation inhibits Smo basolateral accumulation and attenuates Hh target gene expression. In contrast, expression of constitutively active aPKC elevates basolateral accumulation of Smo and promotes Hh signaling. The aPKC-mediated phosphorylation of Smo at Ser680 promotes Ser683 phosphorylation by casein kinase 1 (CK1), and these phosphorylation events elevate Smo activity in vivo. Moreover, aPKC has an additional positive role in Hh signaling by regulating the activity of Cubitus interruptus (Ci) through phosphorylation of the Zn finger DNA-binding domain. Finally, the expression of aPKC is up-regulated by Hh signaling in a Ci-dependent manner. Our findings indicate a direct involvement of aPKC in Hh signaling beyond its role in cell polarity.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Gene Expression Regulation, Enzymologic/physiology , Hedgehog Proteins/metabolism , Protein Kinase C/biosynthesis , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Animals , Casein Kinase I/genetics , Casein Kinase I/metabolism , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Hedgehog Proteins/genetics , Phosphorylation/physiology , Protein Kinase C/genetics , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/genetics , Smoothened Receptor , Transcription Factors/genetics , Up-Regulation/physiology , Wings, Animal/metabolism
20.
Nat Commun ; 4: 2608, 2013.
Article in English | MEDLINE | ID: mdl-24217340

ABSTRACT

Hedgehog (Hh) signalling regulates embryonic development and adult tissue homoeostasis. Mutations of its pathway components including Suppressor of Fused (Sufu) and Gli/Ci predispose to cancers and congenital anomalies. The Sufu-Gli protein complex occupies a central position in the vertebrate Hh signalling pathway, especially in mammals. Here structures of full-length human and Drosophila Sufu, the human Sufu-Gli complex, along with normal mode analysis and FRET measurement results, reveal that Sufu alternates between 'open' and 'closed' conformations. The 'closed' form of Sufu is stabilized by Gli binding and inhibited by Hh treatment, whereas the 'open' state of Sufu is promoted by Gli-dissociation and Hh signalling. Mutations of critical interface residues disrupt the Sufu-Gli complex and prevent Sufu from repressing Gli-mediated transcription, tethering Gli in the cytoplasm and protecting Gli from the 26S proteasome-mediated degradation. Our study thus provides mechanistic insight into the mutual recognition and regulation between Sufu and Gli/Ci.


Subject(s)
Drosophila Proteins/chemistry , Hedgehog Proteins/chemistry , Kruppel-Like Transcription Factors/chemistry , Nuclear Proteins/chemistry , Repressor Proteins/chemistry , Signal Transduction , Transcription Factors/chemistry , Amino Acid Sequence , Animals , Crystallography, X-Ray , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/chemistry , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression Regulation , HEK293 Cells , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kinetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptides/chemical synthesis , Peptides/genetics , Peptides/metabolism , Protein Interaction Domains and Motifs , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Thermodynamics , Transcription Factors/genetics , Transcription Factors/metabolism , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
SELECTION OF CITATIONS
SEARCH DETAIL
...