Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 8(1): 4048, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29511297

ABSTRACT

Esophageal cancer-related gene 4 (Ecrg4) encodes a hormone-like peptide that is believed to be involved in a variety of physiological phenomena, including tumour suppression. Recent progress in the study of Ecrg4 has shown that Ecrg4 is a proinflammatory factor and induces the expression of several cytokines and chemokines in macrophages/microglia. However, the detailed molecular mechanisms of Ecrg4 signalling, especially the Ecrg4 receptors, remain poorly understood. Here, using retrovirus-mediated expression cloning, we identified lectin-like oxidised low-density lipoprotein receptor-1 (LOX-1) as a membrane protein that binds amino acid residues 71-132 of Ecrg4 (Ecrg4(71-132)). Moreover, in addition to LOX-1, several scavenger receptors, such as Scarf1, Cd36 and Stabilin-1, facilitated the efficient internalisation of Ecrg4(71-132) into cells. A broad competitive inhibitor of scavenger receptors, polyinosinic acid, reduced both the binding of Ecrg4(71-132) and the activation of NF-κB in microglia. This activation was dependent on MyD88, an adaptor protein that recruits signalling proteins to Toll-like receptors (TLRs), with the consequent induction of various immune responses. These data suggest that multiple scavenger receptors recognise Ecrg4(71-132) and transduce its signals, together with TLRs, in microglia.


Subject(s)
Microglia/immunology , Neoplasm Proteins/metabolism , Receptors, Scavenger/agonists , Animals , Cell Line , Cytokines/metabolism , Endocytosis , Gene Expression , Genetic Vectors , Humans , Mice , Myeloid Differentiation Factor 88/metabolism , Protein Binding , Rats , Retroviridae/genetics , Signal Transduction
2.
Angew Chem Int Ed Engl ; 55(22): 6452-6, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27079176

ABSTRACT

Targeted transcriptional activation of endogenous genes is important for understanding physiological transcriptional networks, synthesizing genetic circuits, and inducing cellular phenotype changes. The CRISPR/Cas9 system has great potential to achieve this purpose, however, it has not yet been successfully used to efficiently activate endogenous genes and induce changes in cellular phenotype. A powerful method for transcriptional activation by using CRISPR/Cas9 was developed. Replacement of a methylated promoter with an unmethylated one by CRISPR/Cas9 was sufficient to activate the expression of the neural cell gene OLIG2 and the embryonic stem cell gene NANOG in HEK293T cells. Moreover, CRISPR/Cas9-based OLIG2 activation induced the embryonic carcinoma cell line NTERA-2 to express the neuronal marker ßIII-tubulin.


Subject(s)
CRISPR-Cas Systems/genetics , Nanog Homeobox Protein/genetics , Oligodendrocyte Transcription Factor 2/genetics , Cell Line , HEK293 Cells , Humans , Nanog Homeobox Protein/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Transcriptional Activation/genetics
3.
Oncoimmunology ; 5(12): e1242547, 2016.
Article in English | MEDLINE | ID: mdl-28123880

ABSTRACT

Esophageal cancer-related gene 4 (Ecrg4), a hormone-like peptide, is thought to be a tumor suppressor, however, little is known about the mechanism of how Ecrg4 suppresses tumorigenesis. Here, we show that the ecrg4 null glioma-initiating cell (GIC) line, which was generated from neural stem cells of ecrg4 knockout (KO) mice, effectively formed tumors in the brains of immunocompetent mice, whereas the transplanted ecrg4 wild type-GIC line GIC(+/+) was frequently eliminated. This was caused by host immune system including adaptive T cell responses, since depletion of CD4+, CD8+, or NK cells by specific antibodies in vivo recovered tumorigenicity of GIC(+/+). We demonstrate that Ecrg4 fragments, amino acid residues 71-132 and 133-148, which are produced by the proteolitic cleavage, induced the expression of pro-inflammatory cytokines in microglia in vitro. Moreover, blockades of type-I interferon (IFN) signaling in vivo, either depleting IFN-α/ß receptor 1 or using stat1 KO mice, abrogated the Ecrg4-dependent antitumor activity. Together, our findings indicate a major antitumor function of Ecrg4 in enhancing host immunity via type-I IFN signaling, and suggest its potential as a clinical candidate for cancer immunotherapy.

4.
Lung Cancer ; 70(2): 136-45, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20207041

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a critical phenotypic alteration of cancer cells that triggers invasion and metastasis. Lung cancer cells often show mesenchymal phenotypes; however, a causative genetic alteration for the induction of EMT in lung cancer cells remains unknown. Recent studies have shown that the LKB1 gene is mutated in up to one-third of lung adenocarcinomas. Therefore, to pursue the possible involvement of LKB1 inactivation in the induction of EMT in lung carcinogenesis, we generated immortalized lung epithelial cells and lung adenocarcinoma cells with stable or transient LKB1 knockdown. LKB1 knockdown increased cell motility and invasiveness, and induced the expression of several mesenchymal marker proteins accompanied by the expression of ZEB1, a transcriptional repressor for E-cadherin and an EMT inducer. In agreement with the recent findings, expression of miR-200a/c was inversely correlated with that of ZEB1 in LKB1 knockdown clones with mesenchymal phenotype. Furthermore, transient knockdown of LKB1 induced ZEB1 mRNA and increased cell motility, and this motility was suppressed by ZEB1 repression. These results strongly indicate that LKB1 inactivation triggers EMT in lung cancer cells through the induction of ZEB1.


Subject(s)
Adenocarcinoma/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cadherins/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Movement/genetics , Cell Transformation, Neoplastic , Cells, Cultured , Epithelial Cells/pathology , Gene Knockdown Techniques , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/genetics , Mutation/genetics , Neoplasm Invasiveness/genetics , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1
5.
EMBO Rep ; 9(1): 70-5, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18049475

ABSTRACT

WNK kinases are a small group of unique serine/threonine protein kinases that are conserved among multicellular organisms. Mutations in WNK1-4 cause pseudohypoaldosteronism type II-a form of hypertension. WNKs have been linked to the STE20 kinases and ion carriers, but the underlying molecular mechanisms by which WNKs regulate cellular processes in whole animals are unknown. The Caenorhabditis elegans WNK-like kinase WNK-1 interacts with and phosphorylates germinal centre kinase (GCK)-3--a STE20-like kinase--which is known to inactivate CLH-3, a CIC chloride channel. The wnk-1 or gck-3 deletion mutation causes an Exc phenotype, a defect in the tubular extension of excretory canals. Expression of the activated form of GCK-3 or the clh-3 deletion mutation can partly suppress wnk-1 or gck-3 defects, respectively. These results indicate that WNK-1 controls the tubular formation of excretory canals by activating GCK-3, resulting in downregulation of CIC channel activity.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/anatomy & histology , Caenorhabditis elegans/enzymology , Chloride Channels/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans Proteins/genetics , Cell Line , Humans , Models, Biological , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , WNK Lysine-Deficient Protein Kinase 1
6.
Cell Metab ; 5(5): 331-44, 2007 May.
Article in English | MEDLINE | ID: mdl-17488636

ABSTRACT

WNK1 and WNK4 mutations have been reported to cause pseudohypoaldosteronism type II (PHAII), an autosomal-dominant disorder characterized by hyperkalemia and hypertension. To elucidate the molecular pathophysiology of PHAII, we generated Wnk4(D561A/+) knockin mice presenting the phenotypes of PHAII. The knockin mice showed increased apical expression of phosphorylated Na-Cl cotransporter (NCC) in the distal convoluted tubules. Increased phosphorylation of the kinases OSR1 and SPAK was also observed in the knockin mice. Apical localization of the ROMK potassium channel and transepithelial Cl(-) permeability in the cortical collecting ducts were not affected in the knockin mice, whereas activity of epithelial Na(+) channels (ENaC) was increased. This increase, however, was not evident after hydrochlorothiazide treatment, suggesting that the regulation of ENaC was not a genetic but a secondary effect. Thus, the pathogenesis of PHAII caused by a missense mutation of WNK4 was identified to be increased function of NCC through activation of the OSR1/SPAK-NCC phosphorylation cascade.


Subject(s)
Disease Models, Animal , Protein Serine-Threonine Kinases/metabolism , Pseudohypoaldosteronism/physiopathology , Animals , Blood Chemical Analysis , Blood Pressure , Epithelial Sodium Channels/metabolism , Genetic Vectors/genetics , Kidney Tubules, Distal/metabolism , Mice , Microscopy, Fluorescence , Mutation, Missense/genetics , Phosphorylation , Potassium Channels, Inwardly Rectifying/metabolism , Protein Serine-Threonine Kinases/genetics , Sodium Chloride Symporters/metabolism , Urine/chemistry
7.
J Biol Chem ; 280(52): 42685-93, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16263722

ABSTRACT

The WNK1 and WNK4 genes have been found to be mutated in some patients with hyperkalemia and hypertension caused by pseudohypoaldosteronism type II. The clue to the pathophysiology of pseudohypoaldosteronism type II was its striking therapeutic response to thiazide diuretics, which are known to block the sodium chloride cotransporter (NCC). Although this suggests a role for WNK1 in hypertension, the precise molecular mechanisms are largely unknown. Here we have shown that WNK1 phosphorylates and regulates the STE20-related kinases, Ste20-related proline-alanine-rich kinase (SPAK) and oxidative stress response 1 (OSR1). WNK1 was observed to phosphorylate the evolutionary conserved serine residue located outside the kinase domains of SPAK and OSR1, and mutation of the OSR1 serine residue caused enhanced OSR1 kinase activity. In addition, hypotonic stress was shown to activate SPAK and OSR1 and induce phosphorylation of the conserved OSR1 serine residue, suggesting that WNK1 may be an activator of the SPAK and OSR1 kinases. Moreover, SPAK and OSR1 were found to directly phosphorylate the N-terminal regulatory regions of cation-chloride-coupled cotransporters including NKCC1, NKCC2, and NCC. Phosphorylation of NCC was induced by hypotonic stress in cells. These results suggested that WNK1 and SPAK/OSR1 mediate the hypotonic stress signaling pathway to the transporters and may provide insights into the mechanisms by which WNK1 regulates ion balance.


Subject(s)
Cations/chemistry , Nerve Tissue Proteins/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/physiology , Amino Acid Motifs , Amino Acid Sequence , Animals , Biological Transport , Cell Line , Chlorides/chemistry , Cloning, Molecular , Conserved Sequence , Diuretics/pharmacology , Dogs , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Escherichia coli/metabolism , Evolution, Molecular , Glutathione Transferase/metabolism , Humans , Hypertension/pathology , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Ions , Mass Spectrometry , Mice , Minor Histocompatibility Antigens , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Phosphorylation , Protein Kinases/chemistry , Protein Kinases/physiology , Protein Structure, Tertiary , Rats , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid , Serine/chemistry , Signal Transduction , Two-Hybrid System Techniques , WNK Lysine-Deficient Protein Kinase 1
8.
Genes Cells ; 9(6): 549-60, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15189448

ABSTRACT

We have identified and characterized a novel member of the G protein-coupled receptor (GPCR) family, termed DREG. DREG belongs to the LNB-TM7 subfamily and possesses a long amino-terminus that contains a CUB domain, a PTX domain, a hormone binding domain and a GPCR proteolytic site (GPS) domain. RT-PCR experiments and whole mount in situ hybridization in mice showed that DREG is expressed at high levels in the heart and somite during embryogenesis and in the adult lung. When DREG was transiently expressed in mammalian cultured cells, a 35-kD fragment was generated by endogenous proteolytic processing at the conserved GPS domain. This short fragment was found associated with the cell membrane, typical of GPCRs. DREG was further cleaved in the middle of the extracellular domain, generating a soluble 70-kD fragment containing the CUB and PTX domains. This processing was inhibited by an inhibitor of the endoprotease furin but not of matrix metalloproteinases. These results raise the possibility that DREG plays a role in development, not only as a receptor or an adhesion molecule but also as a secreted ligand.


Subject(s)
Conserved Sequence , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Gene Expression Regulation, Developmental , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , COS Cells , Cell Line , Cell Membrane/metabolism , Chlorocebus aethiops , Chromosomes, Human, Pair 6 , Cloning, Molecular , Drosophila Proteins/isolation & purification , Expressed Sequence Tags , Genome, Human , Green Fluorescent Proteins/metabolism , Humans , In Situ Hybridization , Ligands , Mice , Molecular Sequence Data , Open Reading Frames , Plasmids , Point Mutation , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/isolation & purification , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
9.
J Cereb Blood Flow Metab ; 23(9): 1052-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12973021

ABSTRACT

Ischemic tolerance is a phenomenon in which brief episodes of ischemia protect against the lethal effects of subsequent periods of prolonged ischemia. The authors investigated the activation of p38 mitogen-activated protein kinase (p38) in the gerbil hippocampus by Western blotting and immunohistochemistry to clarify the role of p38 kinase in ischemic tolerance. After the 2-minute global ischemia, immunoreactivity indicating active p38 was enhanced at 6 hours of reperfusion and continuously demonstrated 72 hours after ischemia in CA1 and CA3 neurons. Pretreatment with SB203580, an inhibitor of active p38 (0-30 micromol/l), 30 minutes before the 2-minute ischemia reduced the ischemic tolerance effect in a dose-dependent manner. Immunoblot analysis indicated that alteration of the phosphorylation pattern of p38 kinase in the hippocampus after subsequent lethal ischemia was induced by the preconditioning. These findings suggest that lasting activation of p38 may contribute to ischemic tolerance in CA1 neurons of the hippocampus and that components of the p38 cascade can be target molecules to modify neuronal survival after ischemia.


Subject(s)
Brain Ischemia/enzymology , Brain Ischemia/pathology , Hippocampus/enzymology , Ischemic Preconditioning , Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Survival , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gerbillinae , Hippocampus/pathology , Imidazoles/pharmacology , In Situ Nick-End Labeling , MAP Kinase Signaling System/physiology , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Neurons/metabolism , Neurons/pathology , Phosphorylation , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases
10.
EMBO Rep ; 3(1): 69-75, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11751577

ABSTRACT

Recent genetic studies in Drosophila identified a novel non-canonical Wnt pathway, the planar cell polarity (PCP) pathway, that signals via JNK to control epithelial cell polarity in Drosophila. Most recently, a pathway regulating convergent extension movements during gastrulation in vertebrate embryos has been shown to be a vertebrate equivalent of the PCP pathway. However, it is not known whether the JNK pathway functions in this non-canonical Wnt pathway to regulate convergent extension movements in vertebrates. In addition, it is not known whether JNK is in fact activated by Wnt stimulation. Here we show that Wnt5a is capable of activating JNK in cultured cells, and present evidence that the JNK pathway mediates the action of Wnt5a to regulate convergent extension movements in Xenopus. Our results thus demonstrate that the non-canonical Wnt/JNK pathway is conserved in both vertebrate and invertebrate and define that JNK has an activity to regulate morphogenetic cell movements.


Subject(s)
Cell Movement/physiology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases/physiology , Proteins/physiology , Xenopus Proteins , Amino Acid Sequence , Animals , Cloning, Molecular , Drosophila , MAP Kinase Kinase 4 , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Wnt Proteins , Wnt-5a Protein , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...