Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
EMBO Rep ; 20(8): e47052, 2019 08.
Article in English | MEDLINE | ID: mdl-31353791

ABSTRACT

Long non-coding RNAs (lncRNAs) are aberrantly expressed in many disease conditions, including cancer. Accumulating evidence indicates that some lncRNAs may play critical roles in cancer progression and metastasis. Here, we identify a set of lncRNAs that are upregulated in metastatic subpopulations isolated from colon cancer HCT116 cells in vivo and show that one of these lncRNAs, which we name CALIC, is required for the metastatic activity of colon cancer cells. We show that CALIC associates with the RNA-binding protein hnRNP-L and imparts specificity to hnRNP-L-mediated gene expression. Furthermore, we demonstrate that the CALIC/hnRNP-L complex upregulates the tyrosine kinase receptor AXL and that knockdown of CALIC or AXL using shRNA in colon cancer cells attenuates their ability to form metastases in mice. These results suggest that the CALIC/hnRNP-L complex enhances the metastatic potential of colon cancer cells.


Subject(s)
Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Proto-Oncogene Proteins/genetics , RNA, Long Noncoding/genetics , Receptor Protein-Tyrosine Kinases/genetics , Ribonucleoproteins/genetics , Animals , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Disease Progression , Female , HCT116 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Binding , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Ribonucleoproteins/metabolism , Signal Transduction , Survival Analysis , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
2.
Oncol Lett ; 15(3): 4005-4009, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29456745

ABSTRACT

The Wnt signaling pathway is aberrantly activated in the majority of human colorectal tumors. ß-catenin, a key component of the Wnt signaling pathway, interacts with the T-cell factor/lymphoid enhancer-binding factor family of transcription factors and activates transcription of Wnt target genes. Sp5 is one of the Wnt target genes, and its expression is commonly upregulated in colon cancer cells. The present study demonstrates that the expression of Sp5 is not upregulated in the colon cancer cell line HCT116, in which Wnt signaling is constitutively activated. Furthermore, the results demonstrate that Sp5 has the potential to inhibit cell proliferation through upregulation of the cell cycle inhibitor p27. These findings suggest that HCT116 cells downregulate Sp5 to avoid p27-mediated growth arrest.

3.
J Neurosci ; 36(47): 11904-11917, 2016 11 23.
Article in English | MEDLINE | ID: mdl-27881777

ABSTRACT

Lineage development is a stepwise process, governed by stage-specific regulatory factors and associated markers. Astrocytes are one of the principle cell types in the CNS and the stages associated with their development remain very poorly defined. To identify these stages, we performed gene-expression profiling on astrocyte precursor populations in the spinal cord, identifying distinct patterns of gene induction during their development that are strongly correlated with human astrocytes. Validation studies identified a new cohort of astrocyte-associated genes during development and demonstrated their expression in reactive astrocytes in human white matter injury (WMI). Functional studies on one of these genes revealed that mice lacking Asef exhibited impaired astrocyte differentiation during development and repair after WMI, coupled with compromised blood-brain barrier integrity in the adult CNS. These studies have identified distinct stages of astrocyte lineage development associated with human WMI and, together with our functional analysis of Asef, highlight the parallels between astrocyte development and their reactive counterparts associated with injury. SIGNIFICANCE STATEMENT: Astrocytes play a central role in CNS function and associated diseases. Yet the mechanisms that control their development remain poorly defined. Using the developing mouse spinal cord as a model system, we identify molecular changes that occur in developing astrocytes. These molecular signatures are strongly correlated with human astrocyte expression profiles and validation in mouse spinal cord identifies a host of new genes associated with the astrocyte lineage. These genes are present in reactive astrocytes in human white matter injury, and functional studies reveal that one of these genes, Asef, contributes to reactive astrocyte responses after injury. These studies identify distinct stages of astrocyte lineage development and highlight the parallels between astrocyte development and their reactive counterparts associated with injury.


Subject(s)
Astrocytes/metabolism , Astrocytes/pathology , Guanine Nucleotide Exchange Factors/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Spinal Cord Regeneration/physiology , Aging/metabolism , Aging/pathology , Animals , Female , Humans , Male , Mice , Mice, Knockout , Mice, Transgenic , Rho Guanine Nucleotide Exchange Factors , Time Factors
4.
Cell Rep ; 16(10): 2554-2564, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27568568

ABSTRACT

Aberrant activation of Wnt/ß-catenin signaling is a major driving force in colon cancer. Wnt/ß-catenin signaling induces the expression of the transcription factor c-Myc, leading to cell proliferation and tumorigenesis. c-Myc regulates multiple biological processes through its ability to directly modulate gene expression. Here, we identify a direct target of c-Myc, termed MYU, and show that MYU is upregulated in most colon cancers and required for the tumorigenicity of colon cancer cells. Furthermore, we demonstrate that MYU associates with the RNA binding protein hnRNP-K to stabilize CDK6 expression and thereby promotes the G1-S transition of the cell cycle. These results suggest that the MYU/hnRNP-K/CDK6 pathway functions downstream of Wnt/c-Myc signaling and plays a critical role in the proliferation and tumorigenicity of colon cancer cells.


Subject(s)
Cell Cycle , Cyclin-Dependent Kinase 6/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/metabolism , Wnt Signaling Pathway , Animals , Base Sequence , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation , Clone Cells , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Humans , Mice, Nude , RNA, Long Noncoding/genetics , Up-Regulation/genetics
5.
Gastroenterology ; 150(5): 1171-1182, 2016 05.
Article in English | MEDLINE | ID: mdl-26873401

ABSTRACT

BACKGROUND & AIMS: Esophageal squamous cell carcinoma (ESCC) is the predominant form of esophageal cancer in Japan. Smoking and drinking alcohol are environmental risk factors for ESCC, whereas single nucleotide polymorphisms in ADH1B and ALDH2, which increase harmful intermediates produced by drinking alcohol, are genetic risk factors. We conducted a large-scale genomic analysis of ESCCs from patients in Japan to determine the mutational landscape of this cancer. METHODS: We performed whole-exome sequence analysis of tumor and nontumor esophageal tissues collected from 144 patients with ESCC who underwent surgery at 5 hospitals in Japan. We also performed single-nucleotide polymorphism array-based copy number profile and germline genotype analyses of polymorphisms in ADH1B and ALDH2. Polymorphisms in CYP2A6, which increase harmful effects of smoking, were analyzed. Functions of TET2 mutants were evaluated in KYSE410 and HEK293FT cells. RESULTS: A high proportion of mutations in the 144 tumor samples were C to T substitution in CpG dinucleotides (called the CpG signature) and C to G/T substitutions with a flanking 5' thymine (called the APOBEC signature). Based on mutational signatures, patients were assigned to 3 groups, which associated with environmental (drinking and smoking) and genetic (polymorphisms in ALDH2 and CYP2A6) factors. Many tumors contained mutations in genes that regulate the cell cycle (TP53, CCND1, CDKN2A, FBXW7); epigenetic processes (MLL2, EP300, CREBBP, TET2); and the NOTCH (NOTCH1, NOTCH3), WNT (FAT1, YAP1, AJUBA) and receptor-tyrosine kinase-phosphoinositide 3-kinase signaling pathways (PIK3CA, EGFR, ERBB2). Mutations in EP300 and TET2 correlated with shorter survival times, and mutations in ZNF750 associated with an increased number of mutations of the APOBEC signature. Expression of mutant forms of TET2 did not increase cellular levels of 5-hydroxymethylcytosine in HEK293FT cells, whereas knockdown of TET2 increased the invasive activity of KYSE410 ESCC cells. Computational analyses associated the mutations in NFE2L2 we identified with transcriptional activation of its target genes. CONCLUSIONS: We associated environmental and genetic factors with base substitution patterns of somatic mutations and provide a registry of genes and pathways that are disrupted in ESCCs. These findings might be used to design specific treatments for patients with esophageal squamous cancers.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Genomics , Mutation , Polymorphism, Single Nucleotide , Alcohol Dehydrogenase/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Asian People/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/ethnology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , CpG Islands , Cytochrome P-450 CYP2A6/genetics , DNA Mutational Analysis , Esophageal Neoplasms/ethnology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Exome , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene-Environment Interaction , Genetic Association Studies , Genetic Predisposition to Disease , Genomics/methods , HEK293 Cells , Humans , Japan/epidemiology , Oligonucleotide Array Sequence Analysis , Phenotype , Risk Factors , Transfection
6.
Proc Natl Acad Sci U S A ; 113(5): 1273-8, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26768845

ABSTRACT

Many long noncoding RNAs (lncRNAs) are reported to be dysregulated in human cancers and play critical roles in tumor development and progression. Furthermore, it has been reported that many lncRNAs regulate gene expression by recruiting chromatin remodeling complexes to specific genomic loci or by controlling transcriptional or posttranscriptional processes. Here we show that an lncRNA termed UPAT [ubiquitin-like plant homeodomain (PHD) and really interesting new gene (RING) finger domain-containing protein 1 (UHRF1) Protein Associated Transcript] is required for the survival and tumorigenicity of colorectal cancer cells. UPAT interacts with and stabilizes the epigenetic factor UHRF1 by interfering with its ß-transducin repeat-containing protein (TrCP)-mediated ubiquitination. Furthermore, we demonstrate that UHRF1 up-regulates Stearoyl-CoA desaturase 1 and Sprouty 4, which are required for the survival of colon tumor cells. Our study provides evidence for an lncRNA that regulates protein ubiquitination and degradation and thereby plays a critical role in the survival and tumorigenicity of tumor cells. Our results suggest that UPAT and UHRF1 may be promising molecular targets for the therapy of colon cancer.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Colonic Neoplasms/genetics , RNA, Long Noncoding/physiology , CCAAT-Enhancer-Binding Proteins/chemistry , Cell Line, Tumor , Epigenesis, Genetic , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lysine/metabolism , Nerve Tissue Proteins/metabolism , Proteolysis , Ubiquitin-Protein Ligases , Ubiquitination , Up-Regulation
7.
Sci Rep ; 5: 14291, 2015 Sep 21.
Article in English | MEDLINE | ID: mdl-26387746

ABSTRACT

The transcription factor GATA6 is a critical regulator of cell proliferation and development in the gastrointestinal tract. We have recently reported that GATA6 induces the expression of the intestinal stem cell marker LGR5 and enhances the clonogenicity and tumorigenicity of colon cancer cells, but not the growth of these cells cultured under adherent conditions. Here we show that REG4, a member of the regenerating islet-derived (REG) family, is also a target of GATA6. We further demonstrate that REG4 is downregulated by overexpression of miR-363, which suppresses GATA6 expression. Moreover, we show that GATA6-mediated activation of REG4 enhances the growth of colon cancer cells under adherent conditions and is required for their tumorigenicity. Taken together, our findings demonstrate that GATA6 simultaneously induces the expression of genes essential for the growth of colon cancer cells under adherent conditions (REG4) and genes required for their clonogenicity (LGR5), and that the miR-363-GATA6-REG4/LGR5 signaling cascade promotes the tumorigenicity of colon cancer cells.


Subject(s)
Carcinogenesis , Colorectal Neoplasms/genetics , GATA6 Transcription Factor/metabolism , Lectins, C-Type/genetics , Transcriptional Activation , Animals , Carcinogenesis/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/physiopathology , Humans , Mice , Mice, Nude , MicroRNAs/metabolism , Pancreatitis-Associated Proteins , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
8.
Nihon Rinsho ; 73(8): 1283-7, 2015 Aug.
Article in Japanese | MEDLINE | ID: mdl-26281679

ABSTRACT

Tumors are complex tissues composed of cancer cells, various types of stromal cells and extracellular matrix. Recent studies have shown that the tumor microenvironment(TME) plays an important role in both malignant tumor progression and treatment response. Thus, it is essential to improve our understanding of the mechanism of communication between cancer cells and their TME. Much effort has been devoted to developing agents that interfere with the interaction between cancer cells and their TME. In this review, we show a brief overview over the current knowledge about the TME-targeting agents and describe therapeutic strategies that have been designed to target components of the TME.


Subject(s)
Molecular Targeted Therapy , Neoplasms/etiology , Neoplasms/therapy , Tumor Microenvironment , Animals , Disease Progression , Extracellular Matrix/physiology , Fibroblasts/physiology , Humans , Hypoxia , Macrophages/physiology , Neoplasms/blood supply , Neoplasms/metabolism
9.
Lipids Health Dis ; 14: 20, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25884980

ABSTRACT

BACKGROUND: Lactobacillus gasseri SBT2055 (LG2055) has been shown to prevent abdominal adiposity, and suppression of lipid absorption is considered a possible mechanism, detail of which, however, are poorly understood. In the present study, we evaluated the effects of LG2055 on fat hydrolysis by determining pancreatic lipase activity and fat emulsion properties in vitro. We also examined whether LG2055 influences fecal fat excretion in humans. METHODS: Pancreatic lipase activity was investigated in vitro using an artificially prepared fat emulsion and 4-methylumbelliferyl oleate (4-MUO) as substrates. The concentrations of free fatty acids and 4-methylumbelliferone were quantified. Fat emulsion droplet size was measured using a particle size analyzer. The clinical study was performed as a double-blind, randomized, placebo-controlled trial. Subjects consumed 100 g of fermented milk (FM)/d, either with or without LG2055 supplementation, for seven days. Fecal samples were collected during three-day pre-observational and FM intake periods and fecal fat levels were determined. RESULTS: LG2055 dose-dependently suppressed lipase activity in the fat emulsion assay but not in the 4-MUO assay. LG2055 dose-dependently increased fat emulsion droplet size. The effects of LG2055 on lipase activity and fat emulsion properties were increased compared with four other tested strains (Lactobacillus gasseri SBT0317, Lactobacillus gasseri JCM1131T, Lactobacillus. delbrueckii subsp. bulgaricus JCM1002T and Streptococcus thermophilus ATCC19258T). In our clinical study, fecal fat level after FM intake was significantly increased compared with that observed before FM intake in the LG2055-containing active FM group but not the control FM group lacking LG2055. CONCLUSIONS: LG2055 increased fat emulsion droplet size, resulting in the suppression of lipase-mediated fat hydrolysis. The influence of LG2055 on the physicochemical properties of fat emulsion provides a mechanism for the probiotic-mediated suppression of lipid absorption and promotion of fecal fat excretion in humans. TRIAL REGISTRATION: UMIN000015772.


Subject(s)
Fats/metabolism , Fatty Acids/metabolism , Feces/chemistry , Lactobacillus/metabolism , Adult , Aged , Double-Blind Method , Emulsions/metabolism , Fats/analysis , Female , Humans , Hymecromone/analogs & derivatives , Hymecromone/metabolism , Japan , Lipase/metabolism , Male , Middle Aged , Particle Size
10.
Biochem Biophys Res Commun ; 460(2): 216-21, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25770425

ABSTRACT

LGR5 plays an important role in the self-renewal of stem cells and is used as a marker identifying self-renewing stem cells in small intestine and hair follicles. Moreover, LGR5 has been reported to be overexpressed in several cancers. SOX9 is a transcription factor that plays a key role in development, differentiation and lineage commitment in various tissues. It has also been reported that SOX9 is overexpressed in a variety of cancers and contributes to their malignant phenotype. Here we show that LGR5 is required for the tumorigenicity of glioblastoma cells. We further show that SOX9 is upregulated in glioblastoma cells and directly enhances the expression of LGR5. We also demonstrate that knockdown of SOX9 suppresses the proliferation and tumorigenicity of glioblastoma cells. These results suggest that SOX9-mediated transcriptional regulation of LGR5 is critical for the tumorigenicity of glioblastoma cells. We speculate that the SOX9-LGR5 pathway could be a potentially promising target for the therapy of glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Receptors, G-Protein-Coupled/physiology , SOX9 Transcription Factor/physiology , Up-Regulation/physiology , Brain Neoplasms/pathology , Cell Line, Tumor , Gene Knockdown Techniques , Glioblastoma/pathology , Humans , Receptors, G-Protein-Coupled/genetics , SOX9 Transcription Factor/genetics
11.
Biochem Biophys Res Commun ; 459(1): 29-35, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25701787

ABSTRACT

Heterogeneous nuclear ribonucleoprotein K (hnRNP K) is a multi-functional protein involved in transcription, mRNA splicing, mRNA stabilization and translation. Although hnRNP K has been suggested to play a role in the development of many cancers, its molecular function in colorectal cancer has remained elusive. Here we show that hnRNP K plays an important role in the mitotic process in HCT116 colon cancer cells. Furthermore, we demonstrate that hnRNP K directly transactivates the NUF2 gene, the product of which is a component of the NDC80 kinetochore complex and which is known to be critical for a stable spindle microtubule-kinetochore attachment. In addition, knockdown of both hnRNP K and NUF2 caused failure in metaphase chromosome alignment and drastic decrease in the growth of colon cancer cells. These results suggest that the hnRNP K-NUF2 axis is important for the mitotic process and proliferation of colon cancer cells and that this axis could be a target for the therapy of colon cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Colonic Neoplasms/pathology , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Humans , Mice, Inbred BALB C , Mitosis , Promoter Regions, Genetic , Up-Regulation , Xenograft Model Antitumor Assays
12.
J Biol Chem ; 290(7): 4097-109, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25492863

ABSTRACT

Hepatocyte growth factor (HGF) attenuates agonist-induced endothelial cell (EC) permeability and increases pulmonary endothelial barrier function via Rac-dependent enhancement of the peripheral actin cytoskeleton. However, the precise mechanisms of HGF effects on the peripheral cytoskeleton are not well understood. This study evaluated a role for Rac/Cdc42-specific guanine nucleotide exchange factor Asef and the multifunctional Rac effector, IQGAP1, in the mechanism of HGF-induced EC barrier enhancement. HGF induced Asef and IQGAP1 co-localization at the cell cortical area and stimulated formation of an Asef-IQGAP1 functional protein complex. siRNA-induced knockdown of Asef or IQGAP1 attenuated HGF-induced EC barrier enhancement. Asef knockdown attenuated HGF-induced Rac activation and Rac association with IQGAP1, and it abolished both IQGAP1 accumulation at the cell cortical layer and IQGAP1 interaction with actin cytoskeletal regulators cortactin and Arp3. Asef activation state was essential for Asef interaction with IQGAP1 and protein complex accumulation at the cell periphery. In addition to the previously reported role of the IQGAP1 RasGAP-related domain in the Rac-dependent IQGAP1 activation and interaction with its targets, we show that the IQGAP1 C-terminal domain is essential for HGF-induced IQGAP1/Asef interaction and Asef-Rac-dependent activation leading to IQGAP1 interaction with Arp3 and cortactin as a positive feedback mechanism of IQGAP1 activation. These results demonstrate a novel feedback mechanism of HGF-induced endothelial barrier enhancement via Asef/IQGAP1 interactions, which regulate the level of HGF-induced Rac activation and promote cortical cytoskeletal remodeling via IQGAP1-Arp3/cortactin interactions.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Membrane Permeability , Endothelium, Vascular/metabolism , Hepatocyte Growth Factor/pharmacology , Pulmonary Artery/metabolism , ras GTPase-Activating Proteins/metabolism , Blotting, Western , Cells, Cultured , Endothelium, Vascular/cytology , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Pulmonary Artery/cytology , RNA, Small Interfering/genetics , Rho Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , rac GTP-Binding Proteins/metabolism , ras GTPase-Activating Proteins/antagonists & inhibitors , ras GTPase-Activating Proteins/genetics
13.
Mol Biol Cell ; 26(4): 636-50, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25518936

ABSTRACT

Increased levels of hepatocyte growth factor (HGF) in injured lungs may reflect a compensatory response to diminish acute lung injury (ALI). HGF-induced activation of Rac1 GTPase stimulates endothelial barrier protective mechanisms. This study tested the involvement of Rac-specific guanine nucleotide exchange factor Asef in HGF-induced endothelial cell (EC) cytoskeletal dynamics and barrier protection in vitro and in a two-hit model of ALI. HGF induced membrane translocation of Asef and stimulated Asef Rac1-specific nucleotide exchange activity. Expression of constitutively activated Asef mutant mimicked HGF-induced peripheral actin cytoskeleton enhancement. In contrast, siRNA-induced Asef knockdown or expression of dominant-negative Asef attenuated HGF-induced Rac1 activation evaluated by Rac-GTP pull down and FRET assay with Rac1 biosensor. Molecular inhibition of Asef attenuated HGF-induced peripheral accumulation of cortactin, formation of lamellipodia-like structures, and enhancement of VE-cadherin adherens junctions and compromised HGF-protective effect against thrombin-induced RhoA GTPase activation, Rho-dependent cytoskeleton remodeling, and EC permeability. Intravenous HGF injection attenuated lung inflammation and vascular leak in the two-hit model of ALI induced by excessive mechanical ventilation and thrombin signaling peptide TRAP6. This effect was lost in Asef(-/-) mice. This study shows for the first time the role of Asef in HGF-mediated protection against endothelial hyperpermeability and lung injury.


Subject(s)
Cell Membrane Permeability , Guanine Nucleotide Exchange Factors/physiology , Lung/metabolism , Animals , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Hepatocyte Growth Factor/pharmacology , Mice , RNA Interference , Rho Guanine Nucleotide Exchange Factors , Signal Transduction , rac1 GTP-Binding Protein/metabolism
14.
Am J Physiol Lung Cell Mol Physiol ; 308(5): L452-63, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25539852

ABSTRACT

Increased vascular endothelial permeability and inflammation are major pathological mechanisms of pulmonary edema and its life-threatening complication, the acute respiratory distress syndrome (ARDS). We have previously described potent protective effects of hepatocyte growth factor (HGF) against thrombin-induced hyperpermeability and identified the Rac pathway as a key mechanism of HGF-mediated endothelial barrier protection. However, anti-inflammatory effects of HGF are less understood. This study examined effects of HGF on the pulmonary endothelial cell (EC) inflammatory activation and barrier dysfunction caused by the gram-negative bacterial pathogen lipopolysaccharide (LPS). We tested involvement of the novel Rac-specific guanine nucleotide exchange factor Asef in the HGF anti-inflammatory effects. HGF protected the pulmonary EC monolayer against LPS-induced hyperpermeability, disruption of monolayer integrity, activation of NF-kB signaling, expression of adhesion molecules intercellular adhesion molecule-1 and vascular cell adhesion molecule-1, and production of IL-8. These effects were critically dependent on Asef. Small-interfering RNA-induced downregulation of Asef attenuated HGF protective effects against LPS-induced EC barrier failure. Protective effects of HGF against LPS-induced lung inflammation and vascular leak were also diminished in Asef knockout mice. Taken together, these results demonstrate potent anti-inflammatory effects by HGF and delineate a key role of Asef in the mediation of the HGF barrier protective and anti-inflammatory effects. Modulation of Asef activity may have important implications in therapeutic strategies aimed at the treatment of sepsis and acute lung injury/ARDS-induced gram-negative bacterial pathogens.


Subject(s)
Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Guanine Nucleotide Exchange Factors/metabolism , Hepatocyte Growth Factor/pharmacology , Lung Injury/pathology , Lung Injury/physiopathology , Cell Adhesion/drug effects , Cell Membrane Permeability/drug effects , Cell Movement/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Humans , Inflammation/pathology , Lipopolysaccharides , Lung/drug effects , Lung/pathology , Lung/physiopathology , Neutrophils/cytology , Neutrophils/drug effects , Protective Agents/pharmacology , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/drug effects
15.
J Biomater Sci Polym Ed ; 25(9): 895-906, 2014.
Article in English | MEDLINE | ID: mdl-24766535

ABSTRACT

To improve blood compatibility of poly(ether ether ketone) (PEEK), surface modification with methacryloyl-functionalized phospholipid polymers was performed through self-initiation graft polymerization. The copolymers (PMA) of 2-methacryloyloxyethyl phosphorylcholine (MPC) and 2-aminoethyl methacrylate hydrochloride were synthesized by conventional free radical polymerization. The PMA was then immobilized with pentafluorophenyl methacrylate to obtain methacryloyl-functionalized MPC polymers (PMAMA). The degree of substitution of the methacryloyl group into the copolymer was nearly completed. The PMAMA was dissolved in 1-butanol and the solution was dropped on PEEK. UV light (350±50 nm) was subsequently irradiated on PEEK for various periods. Elemental analysis of the PEEK surface was performed by X-ray photoelectron spectroscopy and phosphorus and nitrogen signals due to the MPC units on PEEK were observed. The surface wettability of PEEK was also improved by immobilization of PMAMA. Plasma protein adsorption was effectively reduced on the PMAMA-immobilized surface regardless of the type of protein. Furthermore, PMAMA immobilization was also useful in reducing platelet adhesion on PEEK. In conclusion, methacryloyl-functionalized MPC polymers could be immobilized on PEEK by simple photo-irradiation, resulting in significant improvement in blood compatibility.


Subject(s)
Ketones/chemistry , Methacrylates/chemistry , Phosphorylcholine/analogs & derivatives , Polyethylene Glycols/chemistry , Polymerization , Adsorption , Air , Benzophenones , Blood Proteins/chemistry , Fibrinogen/chemistry , Humans , Materials Testing , Phosphorylcholine/chemistry , Polymers , Surface Properties
16.
Nat Commun ; 5: 3150, 2014.
Article in English | MEDLINE | ID: mdl-24452072

ABSTRACT

Aberrant activation of Wnt signalling results in colorectal tumours. Lgr5 is specifically expressed in stem cells of the intestine and has an essential role in maintaining tissue homeostasis. Lgr5-positive stem cells are responsible for the intestinal adenoma initiated by mutations in adenomatous polyposis coli. Furthermore, Lgr5 interacts with R-spondins and thereby activates Wnt signalling. However, the function of Lgr5 in colorectal tumourigenesis is unclear. Here we show that LGR5 is required for the tumourigenicity of colorectal cancer cells. We show that the transcription factor GATA6 directly enhances the expression of LGR5. We further demonstrate that GATA6 is upregulated in colorectal cancer cells due to the downregulation of miR-363, which directly targets GATA6. Moreover, we show that overexpression of miR-363 suppresses the tumourigenicity of colorectal cancer cells. These results suggest that the miR-363-GATA6-LGR5 pathway is critical for colorectal tumourigenesis and would be a promising target for the treatment of colorectal cancer.


Subject(s)
Carcinogenesis , GATA6 Transcription Factor/metabolism , MicroRNAs/metabolism , Receptors, G-Protein-Coupled/metabolism , Humans
17.
PLoS One ; 8(11): e80609, 2013.
Article in English | MEDLINE | ID: mdl-24244701

ABSTRACT

The tumor suppressor adenomatous polyposis coli (APC) is mutated in sporadic and familial colorectal tumors. APC stimulates the activity of the Cdc42- and Rac1-specific guanine nucleotide exchange factor Asef and promotes the migration and invasion of colorectal tumor cells. Furthermore, Asef is overexpressed in colorectal tumors and is required for colorectal tumorigenesis. It is also known that NOTCH signaling plays critical roles in colorectal tumorigenesis and fate determination of intestinal progenitor cells. Here we show that NOTCH3 up-regulates Asef expression by activating the Asef promoter in colorectal tumor cells. Moreover, we demonstrate that microRNA-1 (miR-1) is down-regulated in colorectal tumors and that miR-1 has the potential to suppress NOTCH3 expression through direct binding to its 3'-UTR region. These results suggest that the miR-1-NOTCH3-Asef pathway is important for colorectal tumor cell migration and may be a promising molecular target for the treatment of colorectal tumors.


Subject(s)
Colorectal Neoplasms/metabolism , MicroRNAs/metabolism , Receptors, Notch/metabolism , 3' Untranslated Regions/genetics , Caco-2 Cells , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Chromatin Immunoprecipitation , Colorectal Neoplasms/genetics , Humans , In Vitro Techniques , MicroRNAs/genetics , Protein Binding , Receptor, Notch3 , Receptors, Notch/genetics , Signal Transduction/genetics , Signal Transduction/physiology
18.
Cancer Sci ; 104(8): 1135-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23910005

ABSTRACT

The tumor suppressor adenomatous polyposis coli (APC) is mutated in familial adenomatous polyposis and in many sporadic colorectal tumors. Adenomatous polyposis coli is known to negatively regulate Wnt signaling by inducing the degradation of ß-catenin. Adenomatous polyposis coli also interacts with the guanine nucleotide exchange factors Asef and Asef2 and stimulates their activity, thereby regulating cell adhesion and migration. Here we show that in confluent, non-motile MDCK II cells, Asef/Asef2 are colocalized with APC at the sites of cell-cell adhesion at the apical and junctional levels. In contrast, in colorectal tumor cells containing mutated APC, significant amounts of Asef/Asef2 and the truncated mutant APCs are localized mainly in the cytoplasm. These results suggest that localization of the Asef/Asef2-APC complex at the sites of cell-cell contact is critical for the regulation of cell adhesion, and that the aberrant subcellular localization of these complexes in colorectal tumor cells may contribute to the cell's aberrant adhesive and migratory properties.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Adenomatous Polyposis Coli/pathology , Animals , Cell Adhesion/physiology , Cell Line, Tumor , Cell Movement/physiology , Cells, Cultured , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Cytoplasm/physiology , Dogs , Madin Darby Canine Kidney Cells , Mutation
19.
J Biol Chem ; 285(2): 1199-207, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19897489

ABSTRACT

Mutation of the tumor suppressor adenomatous polyposis coli (APC) is a key early event in the development of most colorectal tumors. APC promotes degradation of beta-catenin and thereby negatively regulates Wnt signaling, whereas mutated APCs present in colorectal tumor cells are defective in this activity. APC also stimulates the activity of the guanine nucleotide exchange factor Asef and regulates cell morphology and migration. Truncated mutant APCs constitutively activate Asef and induce aberrant migration of colorectal tumor cells. Furthermore, we have recently found that Asef and APC function downstream of hepatocyte growth factor and phosphatidylinositol 3-kinase. We show here that Asef is required for basic fibroblast growth factor- and vascular endothelial growth factor-induced endothelial cell migration. We further demonstrate that Asef is required for basic fibroblast growth factor- and vascular endothelial growth factor-induced microvessel formation. Furthermore, we show that the growth as well as vascularity of subcutaneously implanted tumors are markedly impaired in Asef(-/-) mice compared with wild-type mice. Thus, Asef plays a critical role in tumor angiogenesis and may be a promising target for cancer chemotherapy.


Subject(s)
Adenomatous Polyposis Coli Protein/metabolism , Cell Movement , Colorectal Neoplasms/mortality , Endothelial Cells/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Neovascularization, Pathologic/metabolism , Adenomatous Polyposis Coli Protein/genetics , Animals , Colorectal Neoplasms/genetics , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Guanine Nucleotide Exchange Factors/genetics , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Mice , Mice, Knockout , Neovascularization, Pathologic/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Rho Guanine Nucleotide Exchange Factors , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
20.
EMBO Rep ; 10(12): 1355-62, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19893577

ABSTRACT

Sporadic and familial colorectal tumours usually harbour biallelic adenomatous polyposis coli (APC)-associated mutations that result in constitutive activation of Wnt signalling. Furthermore, APC activates Asef and Asef2, which are guanine-nucleotide exchange factors specific for Rac1 and Cdc42. Here, we show that Asef and Asef2 expression is aberrantly enhanced in intestinal adenomas and tumours. We also show that deficiency of either Asef or Asef2 significantly reduces the number and size of adenomas in Apc(Min/+) mice, which are heterozygous for an APC mutation and spontaneously develop adenomas in the intestine. We observed that the APC-Asef/Asef2 complex induces c-Jun amino-terminal kinase-mediated transactivation of matrix metalloproteinase 9, and is required for the invasive activity of colorectal tumour cells. Furthermore, we show that Asef and Asef2 are required for tumour angiogenesis. These results suggest that Asef and Asef2 have a crucial role in intestinal adenoma formation and tumour progression, and might be promising molecular targets for the treatment of colorectal tumours.


Subject(s)
Adenoma/genetics , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Genes, APC , Guanine Nucleotide Exchange Factors/physiology , Intestinal Neoplasms/genetics , Adenoma/pathology , Animals , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Genes, APC/physiology , Guanine Nucleotide Exchange Factors/genetics , HCT116 Cells , Humans , Intestinal Neoplasms/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness , Rho Guanine Nucleotide Exchange Factors , Signal Transduction/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...