Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Mol Cells ; 45(12): 911-922, 2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36572560

ABSTRACT

A structural protein of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), nucleocapsid (N) protein is phosphorylated by glycogen synthase kinase (GSK)-3 on the serine/arginine (SR) rich motif located in disordered regions. Although phosphorylation by GSK-3ß constitutes a critical event for viral replication, the molecular mechanism underlying N phosphorylation is not well understood. In this study, we found the putative alpha-helix L/FxxxL/AxxRL motif known as the GSK-3 interacting domain (GID), found in many endogenous GSK-3ß binding proteins, such as Axins, FRATs, WWOX, and GSKIP. Indeed, N interacts with GSK-3ß similarly to Axin, and Leu to Glu substitution of the GID abolished the interaction, with loss of N phosphorylation. The N phosphorylation is also required for its structural loading in a virus-like particle (VLP). Compared to other coronaviruses, N of Sarbecovirus lineage including bat RaTG13 harbors a CDK1-primed phosphorylation site and Gly-rich linker for enhanced phosphorylation by GSK-3ß. Furthermore, we found that the S202R mutant found in Delta and R203K/G204R mutant found in the Omicron variant allow increased abundance and hyper-phosphorylation of N. Our observations suggest that GID and mutations for increased phosphorylation in N may have contributed to the evolution of variants.


Subject(s)
Glycogen Synthase Kinase 3 , Nucleocapsid Proteins , SARS-CoV-2 , Humans , Phosphorylation , Nucleocapsid Proteins/genetics
2.
Coron Artery Dis ; 33(8): 643-647, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36302183

ABSTRACT

INTRODUCTION: Drug-eluting stents (DES) significantly improved angiographic and clinical outcomes compared with bare-metal stents in patients with diabetes. The clinical effects of BioMime sirolimus-eluting stent (SES) in patients with diabetes have not been evaluated. Therefore, we compared the efficacy of BioMime DES in coronary artery disease (CAD) patients with versus without diabetes. METHODS: This prospective analytical study compared angiographic in-segment late loss and clinical effectiveness of BioMime SES stents in treating patients with (patients: 77 and lesions: 83) versus without (patients: 154 and lesions: 162) diabetes. The purpose of this study was the comparison of angiographic in-segment late loss at 12 months. Major adverse cardiac events (MACEs) were also monitored as secondary outcomes 24 months after the index procedure. RESULTS: Of 231 patients enrolled in the study, the mean age was 63.3 years and 153 patients were male. Angiographic follow-up rate was 84.8% (patients: 196) and intravascular ultrasound (IVUS) follow-up rate was 67.9% (patients: 157) at 12 months. Diabetic patients were comparable to nondiabetic patients for 12-month in-segment late loss (0.01 ± 0.31 mm for the nondiabetes group versus 0.04 ± 0.11 mm for the diabetes group; P = 0.158; P < 0.05). At 24 months, MACEs, including death, myocardial infarction and ischemic-driven target lesion revascularization were not statistically different between the two treatment groups. CONCLUSIONS: BioMime SES stents in treating patients with diabetes were comparable in reducing angiographic restenosis at 12 months and MACEs at 24 months compared to nondiabetic patients with CAD.


Subject(s)
Coronary Artery Disease , Coronary Restenosis , Diabetes Mellitus , Drug-Eluting Stents , Humans , Male , Middle Aged , Female , Sirolimus/pharmacology , Coronary Restenosis/diagnostic imaging , Coronary Restenosis/etiology , Prospective Studies , Coronary Angiography , Coronary Artery Disease/therapy , Coronary Artery Disease/drug therapy , Stents , Diabetes Mellitus/epidemiology , Diabetes Mellitus/drug therapy , Treatment Outcome
3.
Int J Mol Sci ; 22(17)2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34502497

ABSTRACT

The epithelial-mesenchymal transition (EMT) comprises an important biological mechanism not only for cancer progression but also in the therapeutic resistance of cancer cells. While the importance of the protein abundance of EMT-inducers, such as Snail (SNAI1) and Zeb1 (ZEB1), during EMT progression is clear, the reciprocal interactions between the untranslated regions (UTRs) of EMT-inducers via a competing endogenous RNA (ceRNA) network have received little attention. In this study, we found a synchronized transcript abundance of Snail and Zeb1 mediated by a non-coding RNA network in colorectal cancer (CRC). Importantly, the trans-regulatory ceRNA network in the UTRs of EMT inducers is mediated by competition between tumor suppressive miRNA-34 (miR-34) and miRNA-200 (miR-200). Furthermore, the ceRNA network consisting of the UTRs of EMT inducers and tumor suppressive miRs is functional in the EMT phenotype and therapeutic resistance of colon cancer. In The Cancer Genome Atlas (TCGA) samples, we also found genome-wide ceRNA gene sets regulated by miR-34a and miR-200 in colorectal cancer. These results indicate that the ceRNA networks regulated by the reciprocal interaction between EMT gene UTRs and tumor suppressive miRs are functional in CRC progression and therapeutic resistance.


Subject(s)
Colorectal Neoplasms/metabolism , Genes, Tumor Suppressor , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Neoplasm/metabolism , Snail Family Transcription Factors/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Female , HCT116 Cells , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics , Snail Family Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics
4.
Life Sci Alliance ; 3(7)2020 07.
Article in English | MEDLINE | ID: mdl-32487689

ABSTRACT

Despite the importance of mitochondrial fatty acid oxidation (FAO) in cancer metabolism, the biological mechanisms responsible for the FAO in cancer and therapeutic intervention based on catabolic metabolism are not well defined. In this study, we observe that Snail (SNAI1), a key transcriptional repressor of epithelial-mesenchymal transition, enhances catabolic FAO, allowing pro-survival of breast cancer cells in a starved environment. Mechanistically, Snail suppresses mitochondrial ACC2 (ACACB) by binding to a series of E-boxes located in its proximal promoter, resulting in decreased malonyl-CoA level. Malonyl-CoA being a well-known endogenous inhibitor of fatty acid transporter carnitine palmitoyltransferase 1 (CPT1), the suppression of ACC2 by Snail activates CPT1-dependent FAO, generating ATP and decreasing NADPH consumption. Importantly, combinatorial pharmacologic inhibition of pentose phosphate pathway and FAO with clinically available drugs efficiently reverts Snail-mediated metabolic reprogramming and suppresses in vivo metastatic progression of breast cancer cells. Our observations provide not only a mechanistic link between epithelial-mesenchymal transition and catabolic rewiring but also a novel catabolism-based therapeutic approach for inhibition of cancer progression.


Subject(s)
Acetyl-CoA Carboxylase/genetics , Fatty Acids/metabolism , Genes, Mitochondrial/genetics , Neoplasms/genetics , Neoplasms/metabolism , Oxidation-Reduction , Snail Family Transcription Factors/metabolism , Acetyl-CoA Carboxylase/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , Energy Metabolism , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Humans , Lipid Metabolism , Metabolic Networks and Pathways , Metabolome , Metabolomics , Mice , Neoplasms/pathology
5.
Nat Commun ; 9(1): 2301, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895829

ABSTRACT

Phosphorylation-dependent YAP translocation is a well-known intracellular mechanism of the Hippo pathway; however, the molecular effectors governing YAP cytoplasmic translocation remains undefined. Recent findings indicate that oncogenic YAP paradoxically suppresses Wnt activity. Here, we show that Wnt scaffolding protein Dishevelled (DVL) is responsible for cytosolic translocation of phosphorylated YAP. Mutational inactivation of the nuclear export signal embedded in DVL leads to nuclear YAP retention, with an increase in TEAD transcriptional activity. DVL is also required for YAP subcellular localization induced by E-cadherin, α-catenin, or AMPK activation. Importantly, the nuclear-cytoplasmic trafficking is dependent on the p53-Lats2 or LKB1-AMPK tumor suppressor axes, which determine YAP phosphorylation status. In vivo and clinical data support that the loss of p53 or LKB1 relieves DVL-linked reciprocal inhibition between the Wnt and nuclear YAP activity. Our observations provide mechanistic insights into controlled proliferation coupled with epithelial polarity during development and human cancer.


Subject(s)
Active Transport, Cell Nucleus , Adaptor Proteins, Signal Transducing/metabolism , Dishevelled Proteins/metabolism , Genes, Tumor Suppressor , Phosphoproteins/metabolism , A549 Cells , AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases/metabolism , Animals , Cadherins/metabolism , Cell Cycle Proteins , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Mutational Analysis , Female , HCT116 Cells , HEK293 Cells , Hippo Signaling Pathway , Humans , MCF-7 Cells , Mice , Mice, Nude , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Transcription Factors , Tumor Suppressor Protein p53/metabolism , Wnt Proteins/metabolism , Wnt1 Protein/metabolism , YAP-Signaling Proteins , alpha Catenin/metabolism
6.
J Mol Biol ; 430(12): 1760-1772, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29709570

ABSTRACT

Hsp31 protein, belonging to the DJ-1/ThiJ/PfpI superfamily, increases the survival of Escherichia coli under various stresses. While it was reported as a holding chaperone, Hsp31 was also shown to exhibit the glyoxalase III activity in subsequent study. Here, we describe our finding that Hsp31 undergoes a Zn+2-mediated multimerization (HMWZinc), resulting in an enhanced chaperone activity. Furthermore, it was shown that the formation of HMWZinc is reversible such that the oligomer dissociates into the native dimer by EDTA incubation. We attempted to determine the structural change involving the transition between the native dimer and HMWZinc by adding Ni+2, which is Zn+2-mimetic, producing a potential intermediate structure. An analysis of this intermediate revealed a structure with hydrophobic interior exposed, due to an unfolding of the N-terminal loop and the C-terminal ß-to-α region. A treatment with hydrogen peroxide accelerated HMWZinc formation, so that the Hsp31C185E mutant rendered the formation of HMWZinc even at 45 °C. However, the presence of Zn+2 in the catalytic site antagonizes the oxidation of C185, implying a negative role. Our results suggest an unprecedented mechanism of the enhancing chaperone activity by Hsp31, in which the reversible formation of HMWZinc occurs in the presence of heat and Zn+2 ion.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Zinc/pharmacology , Catalytic Domain , Chromatography, Gel , Escherichia coli/genetics , Escherichia coli Proteins/drug effects , Escherichia coli Proteins/genetics , Hydrogen Peroxide/pharmacology , Models, Molecular , Molecular Chaperones/drug effects , Molecular Chaperones/genetics , Molecular Weight , Mutation , Nickel/pharmacology , Protein Conformation , Protein Multimerization/drug effects , Protein Unfolding
7.
Oncotarget ; 8(19): 31842-31855, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28418862

ABSTRACT

The epithelial-mesenchymal transition (EMT) is implicated in tumorigenesis and cancer progression, and canonical Wnt signaling tightly controls Snail, a key transcriptional repressor of EMT. While the suppression of canonical Wnt signaling and EMT comprises an attractive therapeutic strategy, molecular targets for small molecules reverting Wnt and EMT have not been widely studied. Meanwhile, the anti-helminthic niclosamide has been identified as a potent inhibitor of many oncogenic signaling pathways although its molecular targets have not yet been clearly identified. In this study, we show that niclosamide directly targets Axin-GSK3 interaction, at least in part, resulting in suppression of Wnt/Snail-mediated EMT. In vitro and in vivo, disruption of Axin-GSK3 complex by niclosamide induces mesenchymal to epithelial reversion at nM concentrations, accompanied with suppression of the tumorigenic potential of colon cancer. Niclosamide treatment successfully attenuates Snail abundance while increasing E-cadherin abundance in xenograft tumor. Notably, oral administration of niclosamide significantly suppressed adenoma formation in an APC-MIN mice model, indicating that niclosamide is an effective therapeutic for familial adenomatosis polyposis (FAP) patients. In this study, we identified a novel target to control the canonical Wnt pathway and Snail-mediated EMT program, and discovered a repositioned therapeutics for FAP patients.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Axin Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , Niclosamide/pharmacology , Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/genetics , Animals , Axin Protein/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Glycogen Synthase Kinase 3/chemistry , Heterografts , Mice , Models, Molecular , Molecular Conformation , Niclosamide/chemistry , Protein Binding/drug effects , Wnt Signaling Pathway/drug effects
8.
Nat Commun ; 8: 14374, 2017 02 08.
Article in English | MEDLINE | ID: mdl-28176759

ABSTRACT

Dynamic regulation of glucose flux between aerobic glycolysis and the pentose phosphate pathway (PPP) during epithelial-mesenchymal transition (EMT) is not well-understood. Here we show that Snail (SNAI1), a key transcriptional repressor of EMT, regulates glucose flux toward PPP, allowing cancer cell survival under metabolic stress. Mechanistically, Snail regulates glycolytic activity via repression of phosphofructokinase, platelet (PFKP), a major isoform of cancer-specific phosphofructokinase-1 (PFK-1), an enzyme involving the first rate-limiting step of glycolysis. The suppression of PFKP switches the glucose flux towards PPP, generating NADPH with increased metabolites of oxidative PPP. Functionally, dynamic regulation of PFKP significantly potentiates cancer cell survival under metabolic stress and increases metastatic capacities in vivo. Further, knockdown of PFKP rescues metabolic reprogramming and cell death induced by loss of Snail. Thus, the Snail-PFKP axis plays an important role in cancer cell survival via regulation of glucose flux between glycolysis and PPP.


Subject(s)
Glucose/metabolism , Neoplasms/pathology , Oxidative Stress/genetics , Phosphofructokinase-1, Type C/genetics , Phosphofructokinase-1/genetics , Snail Family Transcription Factors/metabolism , Cell Survival/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycolysis , Humans , NADP/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Pentose Phosphate Pathway/genetics , Phosphofructokinase-1/metabolism , Phosphofructokinase-1, Type C/metabolism , RNA, Small Interfering/metabolism , Snail Family Transcription Factors/genetics
9.
mBio ; 5(5): e01649-14, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25227465

ABSTRACT

UNLABELLED: Cryptococcus neoformans strains isolated from patients with AIDS secrete acid phosphatase, but the identity and role of the enzyme(s) responsible have not been elucidated. By combining a one-dimensional electrophoresis step with mass spectrometry, a canonically secreted acid phosphatase, CNAG_02944 (Aph1), was identified in the secretome of the highly virulent serotype A strain H99. We created an APH1 deletion mutant (Δaph1) and showed that Δaph1-infected Galleria mellonella and mice survived longer than those infected with the wild type (WT), demonstrating that Aph1 contributes to cryptococcal virulence. Phosphate starvation induced APH1 expression and secretion of catalytically active acid phosphatase in the WT, but not in the Δaph1 mutant, indicating that Aph1 is the major extracellular acid phosphatase in C. neoformans and that it is phosphate repressible. DsRed-tagged Aph1 was transported to the fungal cell periphery and vacuoles via endosome-like structures and was enriched in bud necks. A similar pattern of Aph1 localization was observed in cryptococci cocultured with THP-1 monocytes, suggesting that Aph1 is produced during host infection. In contrast to Aph1, but consistent with our previous biochemical data, green fluorescent protein (GFP)-tagged phospholipase B1 (Plb1) was predominantly localized at the cell periphery, with no evidence of endosome-mediated export. Despite use of different intracellular transport routes by Plb1 and Aph1, secretion of both proteins was compromised in a Δsec14-1 mutant. Secretions from the WT, but not from Δaph1, hydrolyzed a range of physiological substrates, including phosphotyrosine, glucose-1-phosphate, ß-glycerol phosphate, AMP, and mannose-6-phosphate, suggesting that the role of Aph1 is to recycle phosphate from macromolecules in cryptococcal vacuoles and to scavenge phosphate from the extracellular environment. IMPORTANCE: Infections with the AIDS-related fungal pathogen Cryptococcus neoformans cause more than 600,000 deaths per year worldwide. Strains of Cryptococcus neoformans isolated from patients with AIDS secrete acid phosphatase; however, the identity and role of the enzyme(s) are unknown. We have analyzed the secretome of the highly virulent serotype A strain H99 and identified Aph1, a canonically secreted acid phosphatase. By creating an APH1 deletion mutant and an Aph1-DsRed-expressing strain, we demonstrate that Aph1 is the major extracellular and vacuolar acid phosphatase in C. neoformans and that it is phosphate repressible. Furthermore, we show that Aph1 is produced in cryptococci during coculture with THP-1 monocytes and contributes to fungal virulence in Galleria mellonella and mouse models of cryptococcosis. Our findings suggest that Aph1 is secreted to the environment to scavenge phosphate from a wide range of physiological substrates and is targeted to vacuoles to recycle phosphate from the expendable macromolecules.


Subject(s)
Acid Phosphatase/genetics , Cryptococcosis/microbiology , Cryptococcus neoformans/enzymology , Fungal Proteins/genetics , Vacuoles/microbiology , Acid Phosphatase/metabolism , Acquired Immunodeficiency Syndrome/microbiology , Animals , Biological Transport , Cryptococcus neoformans/genetics , Disease Models, Animal , Female , Fungal Proteins/metabolism , Gene Deletion , Glucosephosphates/metabolism , Glycerophosphates/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Hydrogen-Ion Concentration , Mannosephosphates/metabolism , Mice , Mice, Inbred BALB C , Monocytes/microbiology , Moths/microbiology , Proteomics , Vacuoles/enzymology
10.
Nat Commun ; 5: 4423, 2014 Jul 23.
Article in English | MEDLINE | ID: mdl-25055241

ABSTRACT

Cytotoxin-associated gene A (CagA) is an oncoprotein and a major virulence factor of H. pylori. CagA is delivered into gastric epithelial cells via a type IV secretion system and causes cellular transformation. The loss of epithelial adhesion that accompanies the epithelial-mesenchymal transition (EMT) is a hallmark of gastric cancer. Although CagA is a causal factor in gastric cancer, the link between CagA and the associated EMT has not been elucidated. Here, we show that CagA induces the EMT by stabilizing Snail, a transcriptional repressor of E-cadherin expression. Mechanistically we show that CagA binds GSK-3 in a manner similar to Axin and causes it to shift to an insoluble fraction, resulting in reduced GSK-3 activity. We also find that the level of Snail protein is increased in H. pylori infected epithelium in clinical samples. These results suggest that H. pylori CagA acts as a pathogenic scaffold protein that induces a Snail-mediated EMT via the depletion of GSK-3.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Proteins/physiology , Down-Regulation/physiology , Epithelial-Mesenchymal Transition/physiology , Glycogen Synthase Kinase 3/metabolism , Helicobacter pylori/physiology , Transcription Factors/physiology , Biopsy , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gastric Mucosa/metabolism , Gastritis/metabolism , Gastritis/pathology , Humans , Signal Transduction/physiology , Snail Family Transcription Factors , Stomach/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
11.
FEBS Lett ; 588(14): 2328-34, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24859084

ABSTRACT

Arginyl-tRNA synthetase (ArgRS) is a tRNA-binding protein that catalyzes the esterification of L-arginine to its cognate tRNA. L-Canavanine, a structural analog of L-arginine, has recently been studied as an anticancer agent. Here, we determined the crystal structures of the apo, L-arginine-complexed, and L-canavanine-complexed forms of the cytoplasmic free isoform of human ArgRS (hArgRS). Similar interactions were formed upon binding to L-canavanine or L-arginine, but the interaction between Tyr312 and the oxygen of the oxyguanidino group was a little bit different. Detailed conformational changes that occur upon substrate binding were explained. The hArgRS structure was also compared with previously reported homologue structures. The results presented here may provide a basis for the design of new anticancer drugs, such as L-canavanine analogs.


Subject(s)
Arginine-tRNA Ligase/chemistry , Amino Acid Sequence , Apoenzymes/chemistry , Arginine/chemistry , Canavanine/chemistry , Catalytic Domain , Crystallography, X-Ray , Humans , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Structural Homology, Protein
12.
Cell Cycle ; 12(10): 1578-87, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23624843

ABSTRACT

p53 is a bona fide tumor suppressor gene whose loss of function marks the most common genetic alteration in human malignancy. Although the causal link between loss of p53 function and tumorigenesis has been clearly demonstrated, the mechanistic links by which loss of p53 potentiates oncogenic signaling are not fully understood. Recent evidence indicates that the microRNA-34 (miR-34) family, a transcriptional target of the p53, directly suppresses a set of canonical Wnt genes and Snail, resulting in p53-mediated suppression of Wnt signaling and the EMT process. In this study, we report that p53 regulates GSK-3ß nuclear localization via miR-34-mediated suppression of Axin2 in colorectal cancer. Exogenous miR-34a decreases Axin2 UTR-reporter activity through multiple binding sites within the 5' and 3' UTR of Axin2. Suppression of Axin2 by p53 or miR-34 increases nuclear GSK-3ß abundance and leads to decreased Snail expression in colorectal cancer cells. Conversely, expression of the non-coding UTR of Axin2 causes depletion of endogenous miR-34 via the miR-sponge effect together with increased Axin2 function, supporting that the RNA-RNA interactions with Axin2 transcripts act as an endogenous decoy for miR-34. Further, RNA transcripts of miR-34 target were correlated with Axin2 in clinical data set of colorectal cancer patients. Although the biological relevance of nuclear GSK-3 level has not been fully studied, our results demonstrate that the tumor suppressor p53/miR-34 axis plays a role in regulating nuclear GSK-3 levels and Wnt signaling through the non-coding UTR of Axin2 in colorectal cancer.


Subject(s)
Axin Protein/metabolism , Glycogen Synthase Kinase 3/metabolism , MicroRNAs/metabolism , Tumor Suppressor Protein p53/metabolism , 3' Untranslated Regions , 5' Untranslated Regions , Antibiotics, Antineoplastic/pharmacology , Axin Protein/antagonists & inhibitors , Axin Protein/genetics , Binding Sites , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Transcription, Genetic , Wnt Signaling Pathway
13.
Sci Signal ; 4(197): ra71, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22045851

ABSTRACT

Although loss of p53 function and activation of canonical Wnt signaling cascades are frequently coupled in cancer, the links between these two pathways remain unclear. We report that p53 transactivated microRNA-34 (miR-34), which consequently suppressed the transcriptional activity of ß-catenin-T cell factor and lymphoid enhancer factor (TCF/LEF) complexes by targeting the untranslated regions (UTRs) of a set of conserved targets in a network of genes encoding elements of the Wnt pathway. Loss of p53 function increased canonical Wnt signaling by alleviating miR-34-specific interactions with target UTRs, and miR-34 depletion relieved p53-mediated Wnt repression. Gene expression signatures reflecting the status of ß-catenin-TCF/LEF transcriptional activity in breast cancer and pediatric neuroblastoma patients were correlated with p53 and miR-34 functional status. Loss of p53 or miR-34 contributed to neoplastic progression by triggering the Wnt-dependent, tissue-invasive activity of colorectal cancer cells. Further, during development, miR-34 interactions with the ß-catenin UTR affected Xenopus body axis polarity and the expression of Wnt-dependent patterning genes. These data provide insight into the mechanisms by which a p53-miR-34 network restrains canonical Wnt signaling cascades in developing organisms and human cancer.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Tumor Suppressor Protein p53/genetics , Wnt Signaling Pathway/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Chick Embryo , Child , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Female , Gene Expression Profiling , HEK293 Cells , Humans , Mice , MicroRNAs/metabolism , Mutation , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA Interference , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Wnt1 Protein/genetics , Wnt1 Protein/metabolism , Xenopus laevis , beta Catenin/genetics , beta Catenin/metabolism
14.
J Cell Biol ; 195(3): 417-33, 2011 Oct 31.
Article in English | MEDLINE | ID: mdl-22024162

ABSTRACT

Snail1 is a zinc finger transcriptional repressor whose pathological expression has been linked to cancer cell epithelial-mesenchymal transition (EMT) programs and the induction of tissue-invasive activity, but pro-oncogenic events capable of regulating Snail1 activity remain largely uncharacterized. Herein, we demonstrate that p53 loss-of-function or mutation promotes cancer cell EMT by de-repressing Snail1 protein expression and activity. In the absence of wild-type p53 function, Snail1-dependent EMT is activated in colon, breast, and lung carcinoma cells as a consequence of a decrease in miRNA-34 levels, which suppress Snail1 activity by binding to highly conserved 3' untranslated regions in Snail1 itself as well as those of key Snail1 regulatory molecules, including ß-catenin, LEF1, and Axin2. Although p53 activity can impact cell cycle regulation, apoptosis, and DNA repair pathways, the EMT and invasion programs initiated by p53 loss of function or mutation are completely dependent on Snail1 expression. These results identify a new link between p53, miR-34, and Snail1 in the regulation of cancer cell EMT programs.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Apoptosis , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Microscopy, Confocal , Neoplasm Invasiveness , Neoplasms/pathology , Snail Family Transcription Factors , Transcription Factors/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , beta Catenin/metabolism
15.
EMBO J ; 29(22): 3787-96, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-20959806

ABSTRACT

Protein O-phosphorylation often occurs reciprocally with O-GlcNAc modification and represents a regulatory principle for proteins. O-phosphorylation of serine by glycogen synthase kinase-3ß on Snail1, a transcriptional repressor of E-cadherin and a key regulator of the epithelial-mesenchymal transition (EMT) programme, results in its proteasomal degradation. We show that by suppressing O-phosphorylation-mediated degradation, O-GlcNAc at serine112 stabilizes Snail1 and thus increases its repressor function, which in turn attenuates E-cadherin mRNA expression. Hyperglycaemic condition enhances O-GlcNAc modification and initiates EMT by transcriptional suppression of E-cadherin through Snail1. Thus, dynamic reciprocal O-phosphorylation and O-GlcNAc modification of Snail1 constitute a molecular link between cellular glucose metabolism and the control of EMT.


Subject(s)
Acetylglucosamine/metabolism , Hyperglycemia/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation , Glucose/metabolism , HEK293 Cells , HeLa Cells , Humans , Molecular Sequence Data , Phosphorylation , Protein Stability , RNA, Messenger/genetics , Serine/metabolism , Snail Family Transcription Factors , Transcription Factors/genetics
16.
Biochem Genet ; 45(7-8): 589-610, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17557203

ABSTRACT

The complete A+T-rich region of mitochondrial DNA has been cloned and sequenced from 48 individuals of Gryllotalpa orientalis (Orthoptera: Gryllotalpidae), collected from five Korean localities. Thirty-six haplotypes acquired from 48 individuals were found to range in size from 917 to 925 bp, with a sequence divergence from 0.1% to 14.3% and an A+T content from 74.5 to 76.3%. Phylogeographic analysis of the G. orientalis haplotypes showed the presence of two clearly differentiated mitochondrial clades, separated by 13.4% of a minimum uncorrected sequence divergence, which suggests the presence of an unknown, similar Gryllotalpa species or a once isolated G. orientalis population. Structural analysis in search of the conserved structural elements previously described in the caeliferan Orthoptera and Diptera revealed that the G. orientalis A+T-rich region harbored a stretch of the [TA(A)]n sequence, which has been suggested to be involved in the control of transcription or replication. In contrast to the abundance of the sequence stretches containing the stem-and-loop structure in the G. orientalis A+T-rich region, the 3' flanking sequence "G(A)nT," which is well conserved in a variety of organisms, including the caeliferan Orthoptera and Diptera, was not conserved in the A+T-rich region of G. orientalis.


Subject(s)
DNA, Mitochondrial/genetics , Gryllidae/genetics , Polymorphism, Genetic , Animals , Base Sequence , Genome, Insect , Haplotypes , Korea , Molecular Sequence Data , Phylogeny , Sequence Alignment
17.
Gene ; 392(1-2): 206-20, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17321076

ABSTRACT

The complete 16,434-bp nucleotide sequence of the mitogenome of the bumble bee, Bombus ignitus (Hymenoptera: Apidae), was determined. The genome contains the base composition and codon usage typical of metazoan mitogenomes. An unusual feature of the B. ignitus mitogenome is the presence of five tRNA-like structures: two each of the tRNALeu(UUR)-like and tRNASer(AGN)-like sequences and one tRNAPhe-like sequence. These tRNA-like sequences have proper folding structures and anticodon sequences, but their functionality in their respective amino acid transfers remained uncertain. Among these sequences, the tRNALeu(UUR)-like sequence and the tRNASer(AGN)-like sequence are seemingly located within the A+T-rich region. This tRNASer(AGN)-like sequence is highly unusual in that its sequence homology is very high compared to the tRNAMet of other insects, including Apis mellifera, but it contains the anticodon ACT, which designates it as tRNASer(AGN). All PCG and rRNAs are conserved in positions observed most frequently in insect mitogenome structures, but the positions of the tRNAs are highly variable, presenting a new arrangement for an insect mitogenome. As a whole, the B. ignitus mitogenome contains the highest A+T content (86.9%) found in any of the complete insects mt sequences determined to date. All protein-coding sequences started with a typical ATN codon. Nine of the 13 PCGs have a complete termination codon (all TAA), but the remaining four genes terminate with the incomplete TA or T. All tRNAs have the typical clover-leaf structures of mt tRNAs, except for tRNASer(AGN), in which the DHU arm forms a simple loop. All anticodons of B. ignitus tRNAs are identical to those of A. mellifera. In the A+T-rich region, a highly conserved sequence block that was previously described in Orthoptera and Diptera was also present. The stem-and-loop structures that may play a role in the initiation of mtDNA replication were also found in this region. Phylogenetic analysis among three corbiculate tribes, represented by Melipona bicolor (Meliponini), A. mellifera (Apini), and B. ignitus (Bombini), showed the closest relationship between M. bicolor and B. ignitus.


Subject(s)
Bees/genetics , Genes, Mitochondrial , Genome, Insect , AT Rich Sequence , Animals , Base Sequence , Female , Molecular Sequence Data , Nucleic Acid Conformation , Open Reading Frames , Phylogeny , RNA, Ribosomal/genetics , RNA, Transfer/genetics
18.
Nat Cell Biol ; 8(12): 1398-406, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17072303

ABSTRACT

Accumulating evidence indicates that hyperactive Wnt signalling occurs in association with the development and progression of human breast cancer. As a consequence of engaging the canonical Wnt pathway, a beta-catenin-T-cell factor (TCF) transcriptional complex is generated, which has been postulated to trigger the epithelial-mesenchymal transition (EMT) that characterizes the tissue-invasive phenotype. However, the molecular mechanisms by which the beta-catenin-TCF complex induces EMT-like programmes remain undefined. Here, we demonstrate that canonical Wnt signalling engages tumour cell dedifferentiation and tissue-invasive activity through an Axin2-dependent pathway that stabilizes the Snail1 zinc-transcription factor, a key regulator of normal and neoplastic EMT programmes. Axin2 regulates EMT by acting as a nucleocytoplasmic chaperone for GSK3beta, the dominant kinase responsible for controlling Snail1 protein turnover and activity. As dysregulated Wnt signalling marks a diverse array of cancerous tissue types, the identification of a beta-catenin-TCF-regulated Axin2-GSK3beta-Snail1 axis provides new mechanistic insights into cancer-associated EMT programmes.


Subject(s)
Breast Neoplasms/pathology , Cytoskeletal Proteins/metabolism , Glycogen Synthase Kinase 3/metabolism , Transcription Factors/metabolism , Wnt Proteins/metabolism , Amino Acid Sequence , Animals , Axin Protein , Breast Neoplasms/genetics , Cell Nucleus/metabolism , Chick Embryo , Cytoplasm/metabolism , Cytoskeletal Proteins/chemistry , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta , Humans , Mesoderm/pathology , Molecular Sequence Data , Neoplasm Invasiveness , Nuclear Export Signals , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Snail Family Transcription Factors , TCF Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Cells, Cultured , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...