Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Epidemiol Health ; 43: e2021052, 2021.
Article in English | MEDLINE | ID: mdl-34412448

ABSTRACT

The Korea National Hospital Discharge In-depth Injury Survey (KNHDIS), which was started in 2005, is a national probability survey of general hospitals in Korea with 100 or more beds conducted by the Korea Disease Control and Prevention Agency (KDCA). The KNHDIS captures approximately 9% of discharged cases from sampled hospitals using a 2-stage stratified cluster sampling scheme, among which 13% are injury related cases, defined as S00-T98 (injury, poisoning, and certain other consequences of external causes) using International Classification of Diseases, 10th revision codes. The KNHDIS collects information on characteristics of injury-related discharges in order to understand the scale of injuries, identify risk factors, and provide data supporting prevention policies and intervention strategies. The types of data captured include the hospitals' information, detailed clinical information, and injury-related codes such as the mechanism, activities undertaken when injured (sports, leisure activities, work, treatment, and education), external causes of the injury, and location of the occurrence of the injury based on the International Classification of External Causes of Injuries. Furthermore, the means of transportation, risk factors for suicide, and toxic substances are recoreded. Annual reports of the KNHDIS are publicly accessible to browse via the KDCA website (http://www.kdca.go.kr) and microdata are available free of charge upon request via email (kcdcinjury@korea.kr).


Subject(s)
Patient Discharge , Wounds and Injuries , Health Care Surveys , Hospitals , Humans , Republic of Korea/epidemiology , Risk Factors , Wounds and Injuries/epidemiology
2.
J Nat Prod ; 82(5): 1325-1330, 2019 05 24.
Article in English | MEDLINE | ID: mdl-30958679

ABSTRACT

The total synthesis of nocarbenzoxazoles F (1) and G (2), originally obtained from the marine-derived halophilic bacterial strain Nocardiopsis lucentensis DSM 44048, was achieved via a simple and versatile route involving microwave-assisted construction of a benzoxazole skeleton, followed by carbon-carbon bond formation with the corresponding aryl bromides. Unfortunately, the 1H and 13C NMR spectra of natural nocarbenzoxazole G did not agree with those of the synthesized compound. In particular, the spectra of the isolated and synthesized compounds showed considerable differences in the signals from the protons and carbons in the aryl group. The revised structure was validated by the total synthesis of the actual nocarbenzoxazole G (8c) molecule, which is a regioisomer of the compound that was reported earlier as nocarbenzoxazole G. The synthesized derivatives showed specific cytotoxicity to the human cervical carcinoma cell line, HeLa, but did not have any remarkable effect on the other cell lines.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Benzoxazoles/chemistry , Benzoxazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , HeLa Cells , Humans , Molecular Structure , Nocardia , Nuclear Magnetic Resonance, Biomolecular
3.
Sci Rep ; 6: 33411, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27651230

ABSTRACT

The dependences of spreading and differentiation of stem cells plated on hydrogel and silicone gel substrates on the rigidity and porosity of the substrates have recently been a subject of some controversy. In experiments on human mesenchymal stem cells plated on soft, medium rigidity, and hard silicone gels we show that harder gels are more osteogenic, softer gels are more adipogenic, and cell spreading areas increase with the silicone gel substrate rigidity. The results of our study indicate that substrate rigidity induces some universal cellular responses independently of the porosity or topography of the substrate.


Subject(s)
Cell Differentiation/drug effects , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Mesenchymal Stem Cells/drug effects , Silicones/pharmacology , Adipogenesis/drug effects , Cell Culture Techniques , Cell Proliferation/drug effects , Humans , Osteogenesis/drug effects , Osteogenesis/genetics
4.
Cancer Res ; 74(23): 6878-89, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25267066

ABSTRACT

CD98hc (SLC3A2) is the heavy chain component of the dimeric transmembrane glycoprotein CD98, which comprises the large neutral amino acid transporter LAT1 (SLC7A5) in cells. Overexpression of CD98hc occurs widely in cancer cells and is associated with poor prognosis clinically, but its exact contributions to tumorigenesis are uncertain. In this study, we showed that genetic deficiency of CD98hc protects against Ras-driven skin carcinogenesis. Deleting CD98hc after tumor induction was also sufficient to cause regression of existing tumors. Investigations into the basis for these effects defined two new functions of CD98hc that contribute to epithelial cancer beyond an intrinsic effect of CD98hc on tumor cell proliferation. First, CD98hc increased the stiffness of the tumor microenvironment. Second, CD98hc amplified the capacity of cells to respond to matrix rigidity, an essential factor in tumor development. Mechanistically, CD98hc mediated this stiffness sensing by increasing Rho kinase (ROCK) activity, resulting in increased transcription mediated by YAP/TAZ, a nuclear relay for mechanical signals. Our results suggest that CD98hc contributes to carcinogenesis by amplifying a positive feedback loop, which increases both extracellular matrix stiffness and resulting cellular responses. This work supports a rationale to explore the use of CD98hc inhibitors as cancer therapeutics.


Subject(s)
Carcinogenesis/metabolism , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Integrins/metabolism , ras Proteins/metabolism , Acyltransferases , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carcinogenesis/pathology , Cell Cycle Proteins , Cell Proliferation/physiology , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Mechanotransduction, Cellular , Mice , Phosphoproteins/metabolism , Signal Transduction/physiology , Skin/metabolism , Skin/pathology , Transcription Factors/metabolism , Tumor Microenvironment/physiology , YAP-Signaling Proteins , rho-Associated Kinases/metabolism
5.
World J Gastroenterol ; 19(26): 4177-84, 2013 Jul 14.
Article in English | MEDLINE | ID: mdl-23864781

ABSTRACT

AIM: To increase satisfaction and diminish anxiety and shame during colonoscopy, we developed novel double pants (NDP) which consist of doubled fabrics with an inner hole. The aim of study was to compare satisfaction, anxiety and shame between NDP and conventional single pants (CSP). METHODS: Total 160 consecutive examinees were randomly divided into NDP and CSP group. Before colonoscopy, questionnaires identifying state and trait anxiety were completed. After colonoscopy, questionnaires for overall satisfaction (Group Health Association of America 9) and pants-specific satisfaction (5-20), state anxiety (20-80), and shame (6-24) were interviewed. RESULTS: Pants-specific satisfaction scores regarding willingness to repeat colonoscopy using same pants (3.3 ± 0.8 vs 2.1 ± 0.9, P < 0.001) and recommendation of same pants to other people (3.3 ± 0.7 vs 2.0 ± 1.0, P < 0.001) were significantly higher in NDP than CSP groups. State anxiety (33.0 ± 7.0 vs 35.4 ± 6.9, P = 0.028) and shame (6.6 ± 1.5 vs 8.1 ± 3.2, P = 0.001) after colonoscopy was lower in NDP group compared with CSP group. CONCLUSION: The NDP contribute to increase satisfaction and decrease anxiety and shame after colonoscopy.


Subject(s)
Clothing , Colonoscopy/instrumentation , Patient Satisfaction , Aged , Anxiety/etiology , Anxiety/prevention & control , Chi-Square Distribution , China , Colonoscopy/adverse effects , Colonoscopy/psychology , Female , Humans , Linear Models , Male , Middle Aged , Multivariate Analysis , Prospective Studies , Shame , Surveys and Questionnaires
6.
Gut Liver ; 7(1): 51-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23423645

ABSTRACT

BACKGROUND/AIMS: The symptoms of inflammatory bowel disease (IBD) fluctuate considerably over time. However, it has not been determined whether these symptoms are affected by the menstrual cycle in female IBD patients. This study analyzed the effects of the menstrual cycle on IBD symptom variation. METHODS: This was a prospective study of 91 study subjects (47 IBD patients and 44 healthy controls) who reported daily symptoms and signs throughout their menstrual cycles. RESULTS: IBD patients had significantly more frequent gastrointestinal symptoms, such as nausea (30% vs 7%, p=0.006), flatulence (53% vs 22%, p=0.003), and abdominal pain as compared to controls (68% vs 38%, p=0.006). The IBD patients also experienced more frequent systemic premenstrual symptoms than the controls (79% vs 50%, p=0.003). More severe abdominal pain (p=0.002) and lower mean general condition scores (p=0.001) were noted during the menstrual phase as compared to the pre- or post-menstrual phase in both groups. IBD patients experienced more frequent premenstrual gastrointestinal symptoms than controls, but their IBD symptoms did not change significantly during the menstrual cycle. CONCLUSIONS: Knowledge of the cyclic alterations in gastrointestinal and systemic symptoms may be helpful in determining the true exacerbation of disease in female IBD patients.

7.
J Cell Sci ; 125(Pt 24): 5960-73, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23077174

ABSTRACT

Transmembrane 4 L six family member 5 (TM4SF5) plays an important role in cell migration, and focal adhesion kinase (FAK) activity is essential for homeostatic and pathological migration of adherent cells. However, it is unclear how TM4SF5 signaling mediates the activation of cellular migration machinery, and how FAK is activated during cell adhesion. Here, we showed that direct and adhesion-dependent binding of TM4SF5 to FAK causes a structural alteration that may release the inhibitory intramolecular interaction in FAK. In turn, this may activate FAK at the cell's leading edge, to promote migration/invasion and in vivo metastasis. TM4SF5-mediated FAK activation occurred during integrin-mediated cell adhesion. TM4SF5 was localized at the leading edge of the cells, together with FAK and actin-organizing molecules, indicating a signaling link between TM4SF5/FAK and actin reorganization machinery. Impaired interactions between TM4SF5 and FAK resulted in an attenuated FAK phosphorylation (the signaling link to actin organization machinery) and the metastatic potential. Our findings demonstrate that TM4SF5 directly binds to and activates FAK in an adhesion-dependent manner, to regulate cell migration and invasion, suggesting that TM4SF5 is a promising target in the treatment of metastatic cancer.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Focal Adhesion Kinase 1/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Tetraspanins/genetics , Amino Acid Sequence , Animals , Carcinoma, Hepatocellular/enzymology , Cell Adhesion/physiology , Cell Movement/physiology , Enzyme Activation , Female , Heterografts , Humans , Liver Neoplasms/enzymology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasm Metastasis , Phosphorylation , Signal Transduction , Tetraspanins/metabolism
8.
Biochem J ; 443(3): 691-700, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22292774

ABSTRACT

The EMT (epithelial-mesenchymal transition) is involved in fibrosis and cancer, and is regulated by different signalling pathways mediated through soluble factors, actin reorganization and transcription factor actions. Because the tetraspan (also called tetraspanin) TM4SF5 (transmembrane 4 L6 family member 5) is highly expressed in hepatocellular carcinoma and induces EMT, understanding how TM4SF5 expression in hepatocytes is regulated is important. We explored the mechanisms that induce TM4SF5 expression and whether impaired signalling pathways for TM4SF5 expression inhibit the acquisition of mesenchymal cell features, using human and mouse normal hepatocytes. We found that TGFß1 (transforming growth factor ß1)-mediated Smad activation caused TM4SF5 expression and EMT, and activation of the EGFR [EGF (epidermal growth factor) receptor] pathway. Inhibition of EGFR activity following TGFß1 treatment abolished acquisition of EMT, suggesting a link from Smads to EGFR for TM4SF5 expression. Further, TGFß1-mediated EGFR activation and TM4SF5 expression were abolished by EGFR suppression or extracellular EGF depletion. Smad overexpression mediated EGFR activation and TM4SF5 expression in the absence of serum, and EGFR kinase inactivation or EGF depletion abolished Smad-overexpression-induced TM4SF5 and mesenchymal cell marker expression. Inhibition of Smad, EGFR or TM4SF5 using Smad7 or small compounds also blocked TM4SF5 expression and/or EMT. These results indicate that TGFß1- and growth factor-mediated signalling activities mediate TM4SF5 expression leading to acquisition of mesenchymal cell features, suggesting that TM4SF5 induction may be involved in the development of liver pathologies.


Subject(s)
Epithelial-Mesenchymal Transition , ErbB Receptors/metabolism , Membrane Proteins/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism , Cell Line, Tumor , Humans
9.
J Cell Biochem ; 112(3): 782-92, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21328452

ABSTRACT

Transmembrane 4 L six family member 5 (TM4SF5) is highly expressed in hepatocarcinoma and causes epithelial-mesenchymal transition (EMT) of hepatocytes. We found that TM4SF5-expressing cells showed lower mRNA levels but maintained normal protein levels in certain gene cases, indicating that TM4SF5 mediates stabilization of proteins. In this study, we explored whether regulation of proteasome activity and TM4SF5 expression led to EMT. We observed that TM4SF5 expression caused inhibition of proteasome activity and proteasome subunit expression, causing morphological changes and loss of cell-cell contacts. shRNA against TM4SF5 recovered proteasome expression, with leading to blockade of proteasome inactivation and EMT. Altogether, TM4SF5 expression appeared to cause loss of cell-cell adhesions via proteasome suppression and thereby proteasome inhibition, leading to repression of cell-cell adhesion molecules, such as E-cadherin.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Membrane Proteins/biosynthesis , Proteasome Inhibitors , Recombinant Proteins/biosynthesis , Cadherins/metabolism , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Down-Regulation , Enzyme Assays , Hepatocyte Growth Factor/pharmacology , Humans , Leupeptins/pharmacology , Membrane Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , RNA/metabolism , RNA Interference , Recombinant Proteins/genetics , Transcription, Genetic , Ubiquitination , beta Catenin/metabolism
10.
Front Biosci (Landmark Ed) ; 16(5): 1752-8, 2011 01 01.
Article in English | MEDLINE | ID: mdl-21196261

ABSTRACT

TM4SF5 is a transmembrane glycoprotein of the transmembrane 4 L six family, a branch of the tetraspanin family and highly expressed in many types of cancers. TM4SF5 induces epithelial-mesenchymal transition (EMT) by morphological changes resulting from inactivation of RhoA mediated by stabilized cytosolic p27kip1. TM4SF5-mediated EMT can lead to loss of contact inhibition and enhanced migration/invasion, presumably depending on cross-talks between TM4SF5 and integrins. An anti-TM4SF5 agent appears to target the second extracellular domain of TM4SF5, which is important for cross-talk with integrins, leading to a blockade of TM4SF5-mediated multilayer growth and migration/invasion. In addition, TM4SF5 engages in cross-talk with integrin alpha5 to induce and secrete VEGF, which in turn causes activation of angiogenesis in endothelial cells. Therefore, TM4SF5 plays a central regulatory role in a wide variety of physiological processes through cross-talk with integrins. This review presents current knowledge from in vitro and in vivo observations of the roles of TM4SF5-integrin cooperation in hepatocellular carcinogenesis and discusses important areas for future investigation.


Subject(s)
Integrins/physiology , Membrane Proteins/physiology , Actins/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Chalcone/analogs & derivatives , Chalcone/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/drug effects , Neoplasm Invasiveness/physiopathology , Neovascularization, Pathologic/chemically induced , Sulfonamides/pharmacology , Tumor Microenvironment/drug effects
11.
Biochimie ; 93(2): 168-74, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20888885

ABSTRACT

In this study, we demonstrate that raloxifene, a selective estrogen receptor modulator, is a potent inducer of the anti-inflammatory enzyme heme oxygenase-1 (HO-1). In RAW264.7 macrophages, raloxifene induced HO-1 mRNA and protein expression. Pretreatment of ICI182780, an estrogen receptor (ER) antagonist or knock-down of endogenous ERα or ERß gene by RNA interference failed to reverse raloxifene-mediated HO-1 induction, indicating an estrogen receptor-independent mechanism. Interestingly, the raloxifene-induced HO-1 expression was suppressed by reactive oxygen species (ROS) scavengers, including glutathione, TEMPO, Me(2)SO, 1,10-phenanthroline, or allopurinol. In addition, buthionine sulfoximine, an inhibitor of reduced glutathione synthesis, or Fe(2+)/Cu(2+) ions enhanced the positive effect of raloxifene on HO-1 expression. Consistent with these findings, raloxifene induced production of intracellular ROS and increased xanthine oxidase activity in vitro. Additional experiments revealed the involvement of mitogen-activated protein kinase (MAPK) kinase6 and p38 MAPK in the up-regulation of HO-1 by raloxifene and identified p38 MAPK as a downstream effector of ROS. Furthermore, the ROS-p38 MAPK cascade targeted the transcription factor cAMP-responsive element-binding protein (CREB). Finally, the functional significance of HO-1 induction was revealed by raloxifene-mediated inhibition of inducible nitric oxide synthase expression and nitric oxide production, a response reversed by the inhibition of HO-1 protein synthesis or blockade of p38 MAPK or xanthine oxidase activity. Therefore, identification of ROS-p38 MAPK-CREB-linked cascade as cellular relays in raloxifene-mediated HO-1 expression defines the signaling events that could participate in raloxifene-mediated anti-inflammatory response.


Subject(s)
Heme Oxygenase-1/metabolism , Lipopolysaccharides/pharmacology , Nitric Oxide/biosynthesis , Raloxifene Hydrochloride/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/biosynthesis , Hydrogen Peroxide/metabolism , Iron/metabolism , Macrophages/drug effects , Macrophages/enzymology , Macrophages/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Xanthine/metabolism , Xanthine Oxidase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Cancer Biol Ther ; 11(3): 330-6, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21099346

ABSTRACT

Two separate clinical studies of advanced hepatocarcinoma patients recently reported that the multikinase inhibitor sorafenib (nexavar) could extend survival of the patients only by 2-3 months. We also previously demonstrated that 4'-(p-toluenesulfonylamido)-4-hydroxychalcone (TSAHC) blocks the multilayer growth and migration mediated by TM4SF5, which is highly expressed in approximately 80% of Korean hepatocarcinoma patients. Therefore, we wondered how TSAHC might be different from sorafenib to deal with hepatocarcinoma in terms of the therapeutic characteristics including specificity for TM4SF5. TM4SF5 is previously shown to mediate tumorigenesis through cytosolic p27Kip1-mediated inactivation of RhoA, epithelial-mesenchymal transition, multilayer growth, migration, invasion, and tumor angiogenesis. In this study, TSAHC and two derivatives showed similar antagonistic activities against TM4SF5-mediated signaling and multilayer growth in vitro and anti-tumorigenic activity even in early stages of TM4SF5-mediated tumor formation in nude mice. Meanwhile, sorafenib was only effective much later in tumorigenesis in vivo and affected in vitro proliferation in a TM4SF5-independent manner. Altogether, these observations suggest that TSAHC may be a promising anti-tumorigenic reagent, especially against TM4SF5-mediated hepatocarcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Benzenesulfonates/pharmacology , Carcinoma, Hepatocellular/drug therapy , Chalcone/analogs & derivatives , Liver Neoplasms/drug therapy , Membrane Proteins/metabolism , Pyridines/pharmacology , Sulfonamides/pharmacology , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chalcone/metabolism , Chalcone/pharmacology , Drug Discovery , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Proteins/genetics , Mice , Mice, Nude , Niacinamide/analogs & derivatives , Phenylurea Compounds , Signal Transduction , Sorafenib , Sulfonamides/metabolism , Xenograft Model Antitumor Assays
13.
J Biol Chem ; 285(46): 36021-31, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20829364

ABSTRACT

Protein-protein interactions and/or signaling activities at focal adhesions, where integrin-mediated adhesion to extracellular matrix occurs, are critical for the regulation of adhesion-dependent cellular functions. Although the phosphorylation and activities of focal adhesion molecules have been intensively studied, the effects of the O-GlcNAc modification of their Ser/Thr residues on cellular functions have been largely unexplored. We investigated the effects of O-GlcNAc modification on actin reorganization and morphology of rat insulinoma INS-1 cells after glucosamine (GlcN) treatment. We found that paxillin, a key adaptor molecule in focal adhesions, could be modified by O-GlcNAc in INS-1 cells treated with GlcN and in pancreatic islets from mice treated with streptozotocin. Ser-84/85 in human paxillin appeared to be modified by O-GlcNAc, which was inversely correlated to Ser-85 phosphorylation (Ser-83 in rat paxillin). Integrin-mediated adhesion signaling inhibited the GlcN treatment-enhanced O-GlcNAc modification of paxillin. Adherent INS-1 cells treated with GlcN showed restricted protrusions, whereas untreated cells showed active protrusions for multiple-elongated morphologies. Upon GlcN treatment, expression of a triple mutation (S83A/S84A/S85A) resulted in no further restriction of protrusions. Together these observations suggest that murine pancreatic ß cells may have restricted actin organization upon GlcN treatment by virtue of the O-GlcNAc modification of paxillin, which can be antagonized by a persistent cell adhesion process.


Subject(s)
Acetylglucosamine/metabolism , Actins/metabolism , Glucosamine/pharmacology , Paxillin/metabolism , Animals , Blotting, Western , Cell Adhesion/drug effects , Cell Line, Tumor , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Humans , Immunohistochemistry , Insulinoma/metabolism , Insulinoma/pathology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Laminin/metabolism , Mice , Mice, Inbred ICR , Microscopy, Fluorescence , Mutation , Paxillin/genetics , Phosphorylation/drug effects , Rats , Serine/genetics , Serine/metabolism , Streptozocin
14.
J Cell Biochem ; 111(1): 59-66, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20506553

ABSTRACT

Overexpression of transmembrane 4 L six family member 5 (TM4SF5), a four-transmembrane L6 family member, causes aberrant cell proliferation and angiogenesis, but the roles of TM4SF5 in migration, invasion, and tumor metastasis remain unknown. Using in vitro hepatocarcinoma cells that ectopically or endogenously express TM4SF5 and in vivo mouse systems, roles of TM4SF5 in metastatic potentials were examined. We found that TM4SF5 expression facilitated migration, invadopodia formation, MMP activation, invasion, and eventually lung metastasis in nude mice, but suppression of TM4SF5 with its shRNA blocked the effects. Altogether, TM4SF5-mediated migration and invasion suggest that TM4SF5 may be therapeutically targeted to deal with TM4SF5-mediated hepatocellular cancers.


Subject(s)
Carcinoma, Hepatocellular , Cell Movement/physiology , Hepatocytes , Liver Neoplasms , Membrane Proteins/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Female , Hepatocytes/cytology , Hepatocytes/pathology , Hepatocytes/physiology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis
15.
Biochim Biophys Acta ; 1803(8): 975-82, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20399237

ABSTRACT

Transmembrane 4 L six family member 5 (TM4SF5) causes epithelial-mesenchymal transition (EMT) for aberrant cell proliferation. However, the effects of TM4SF5 expression on cell cycle are unknown so far. In this study, using hepatocytes that either ectopically or endogenously express TM4SF5 and human hepatocarcinoma tissues, the role of TM4SF5 in G1/S phase progression was examined. We found that TM4SF5 expression accelerated G1/S phase progression with facilitated cyclin D1 and E expression and Rb phosphorylation. Furthermore, TM4SF5 enhanced trafficking of CDK4 and cyclin D1 into the nucleus and induced complex formation between them. However, TM4SF5-facilitated G1/S phase progression was blocked by silencing of p27Kip1 using siRNA or by infection of active RhoA. Pharmacological inhibition of ROCK accelerated the G1/S phase progression of control TM4SF5-unexpressing cells. Altogether, these observations suggest that TM4SF5 accelerates G1/S phase progression with facilitated CDK4/cyclin D1 entry into the nucleus, which might be supported by TM4SF5-mediated actin reorganization through cytosolic p27Kip1 expression and Rho GTPase activity.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , G1 Phase/physiology , Membrane Proteins/metabolism , S Phase/physiology , rhoA GTP-Binding Protein/metabolism , Cell Line, Tumor , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Membrane Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/genetics
16.
Carcinogenesis ; 30(11): 1872-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19789264

ABSTRACT

Four-transmembrane L6 family member 5 (TM4SF5) and its homolog L6, a tumor antigen, form a four-transmembrane L6 family. TM4SF5 expression causes uncontrolled cell proliferation and angiogenesis. Although other genuine transmembrane 4 superfamily (TM4SF) members co-operate with integrins for cell migration, roles of TM4SF5 in the cellular spreading and migration are unknown. Using hepatocarcinoma cell clones that ectopically express TM4SF5, we found that cross talks via an extracellular interaction between TM4SF5 and integrin alpha2 in collagen type I environment inhibited integrin alpha2 functions such as spreading on and migration toward collagen I, which were recovered by suppression of TM4SF5 or structural disturbance of its second extracellular loop using a peptide or mutagenesis. Altogether, the observations suggest that TM4SF5 in hepatocytes negatively regulates integrin alpha2 function via an interaction between the extracellular loop 2 of TM4SF5 and integrin alpha2 during cell spreading on and migration through collagen I environment.


Subject(s)
Integrin alpha2/metabolism , Liver Neoplasms/genetics , Membrane Proteins/metabolism , Antigens, Neoplasm/metabolism , Cell Movement , Clone Cells , Collagen Type I/metabolism , Extracellular Matrix , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Integrin alpha2/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Proteins/genetics , Neoplasm Invasiveness/genetics , Protein Binding , Protein Structure, Tertiary , Receptor Cross-Talk
17.
Korean J Gastroenterol ; 54(2): 126-9, 2009 Aug.
Article in Korean | MEDLINE | ID: mdl-19696541

ABSTRACT

The gastrointestinal (GI) tract is the most frequently involved site of mucosa-associated lymphoid tissue (MALT) lymphoma. Stomach is the most common site of involvement among the GI tract. However, MALT lymphoma of the large intestine is rare. A diagnosis is established by pathological examination of the surgical or endoscopic specimens. A 72-year-old man with low abdominal pain was diagnosed as a sigmoid MALT lymphoma, which was noted as an obstructing mass in a colonoscopic examination. A left hemicolectomy was performed, and the patient has had no recurrence postoperatively without any chemotherapy.


Subject(s)
Colon, Sigmoid/pathology , Lymphoma, B-Cell, Marginal Zone/diagnosis , Aged , Colonoscopy , Diagnosis, Differential , Humans , Lymphoma, B-Cell, Marginal Zone/diagnostic imaging , Lymphoma, B-Cell, Marginal Zone/pathology , Male , Radiography
18.
Biochim Biophys Acta ; 1793(5): 781-91, 2009 May.
Article in English | MEDLINE | ID: mdl-19339212

ABSTRACT

Cell adhesion to the extracellular matrix (ECM) can activate signaling via focal adhesion kinase (FAK) leading to dynamic regulation of cellular morphology. Mechanistic basis for the lack of effective intracellular signaling by non-attached epithelial cells is poorly understood. To examine whether signaling in suspended cells is regulated by Fer cytoplasmic tyrosine kinase, we investigated the effect of ectopic Fer expression on signaling in suspended or adherent hepatocytes. We found that ectopic Fer expression in Huh7 hepatocytes in suspension or on non-permissive poly-lysine caused significant phosphorylation of FAK Tyr577, Tyr861, or Tyr925, but not Tyr397 or Tyr576. Fer-mediated FAK phosphorylation in suspended cells was independent of c-Src activity or growth factor stimulation, but dependent of cortactin expression. Consistent with these results, complex formation between FAK, Fer, and cortactin was observed in suspended cells. The Fer-mediated effect correlated with multiple membrane protrusions, even on poly-lysine. Together, these observations suggest that Fer may allow a bypass of anchorage-dependency for intracellular signal transduction in hepatocytes.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Hepatocytes/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Tyrosine/metabolism , Animals , Cell Adhesion/physiology , Cell Culture Techniques , Cell Membrane/metabolism , Cell Surface Extensions/metabolism , Cells, Cultured , Cortactin/genetics , Cortactin/metabolism , Epidermal Growth Factor/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Hepatocytes/cytology , Humans , Phosphorylation , Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , src-Family Kinases/metabolism
19.
Hepatology ; 49(4): 1316-25, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19177595

ABSTRACT

UNLABELLED: We previously reported that the four-transmembrane L6 family member 5 (TM4SF5) was highly expressed in hepatocarcinoma, induced morphological elongation and epithelial-mesenchymal transition, and caused abnormal cell growth in multilayers in vitro and tumor formation in vivo. In this study, we identified a synthetic compound, 4'-(p-toluenesulfonylamido)-4-hydroxychalcone (TSAHC) that antagonized both the TM4SF5-mediated multilayer growth and TM4SF5-enhanced migration/invasion. TSAHC treatment induced multilayer-growing cells to grow in monolayers, recovering contact inhibition without accompanying apoptosis, and inhibited chemotactic migration and invasion. Tumor formation in nude mice injected with TM4SF5-expressing cells and the growth of cells expressing endogenous TM4SF5, but not of TM4SF5-null cells, was suppressed by treatment with TSAHC, but not by treatment with its analogs. The structure-activity relationship indicated the significance of 4'-p-toluenesulfonylamido and 4-hydroxy groups for the anti-TM4SF5 effects of TSAHC. Point mutations of the putative N-glycosylation sites abolished the TM4SF5-specific TSAHC responsiveness. CONCLUSION: These observations suggest that TM4SF5-enhanced tumorigenic proliferation and metastatic potential can be blocked by TSAHC, likely through targeting the extracellular region of TM4SF5, which is important for protein-protein interactions.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Chalcone/analogs & derivatives , Chalcones/pharmacology , Hepatocytes/metabolism , Liver Neoplasms/metabolism , Membrane Proteins/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Carcinogenicity Tests , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Chalcone/pharmacology , Contact Inhibition/drug effects , Glycosylation , Humans , Membrane Proteins/genetics , Mice , Phenotype , Point Mutation , Protein Interaction Domains and Motifs , Structure-Activity Relationship
20.
Blood ; 113(8): 1845-55, 2009 Feb 19.
Article in English | MEDLINE | ID: mdl-19036703

ABSTRACT

Tetraspan TM4SF5 is highly expressed in a diverse number of tumor types. Here we explore the mechanistic roles of TM4SF5 in angiogenesis. We found that TM4SF5 overexpression correlates with vascular endothelial growth factor (VEGF) expression in SNU449 hepatocytes and with vessel formation in clinical hepatocarcinoma samples. Conditioned media from TM4SF5-expressing cells enhanced viability and tube formation of primary human umbilical vein endothelial cells, and outgrowth of endothelial cells from aorta ring segments, which was abolished by treatment with an anti-VEGF antibody. TM4SF5 retained integrin alpha(5) on the cell surface for VEGF induction, and preincubation with anti-integrin alpha(5) antibody abolished TM4SF5-mediated VEGF expression and secretion. TM4SF5-mediated effects required integrin alpha(5), c-Src, and signal transducer and activator of transcription 3 (STAT3). In addition, tumors from nude mice injected with TM4SF5-expressing cells and from clinical human hepatocarcinoma tissues showed enhanced integrin alpha(5) expression, vessel formation, and signaling activity, which were inhibited by administration of anti-integrin alpha(5) or -VEGF antibody. This study suggests that TM4SF5 facilitates angiogenesis of neighboring endothelial cells through VEGF induction, mediated by cooperation between TM4SF5 and integrin alpha(5) of epithelial cells.


Subject(s)
Carcinoma, Hepatocellular/blood supply , Integrin alpha5/metabolism , Liver Neoplasms/blood supply , Membrane Proteins/metabolism , Neovascularization, Physiologic/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Antibodies/pharmacology , Aorta/cytology , CSK Tyrosine-Protein Kinase , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Culture Media, Conditioned/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Integrin alpha5/immunology , Liver Neoplasms/metabolism , Male , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Transfection , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/immunology , src-Family Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...