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










Publication year range
1.
Heliyon ; 10(2): e24425, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38293488

ABSTRACT

Electronic textile-based gas sensors with a high response for NO2 gas were fabricated using reduced graphene oxide (rGO)-coated commercial cotton fabric (rGOC). Graphene oxide (GO) was coated on cotton fabric by simply dipping the cotton into a GO solution. To investigate the relationship between the degree of reduction and the sensing response, the GO-coated fabrics were thermally reduced at various temperatures (190, 200, 300, and 400 °C). The change in the amount of oxygen functional groups on the rGOCs was observed by x-ray photoelectron spectroscopy, Raman spectroscopy, and x-ray diffraction patterns. The maximum sensing response of 45.90 % at 10 ppm of NO2 gas at room temperature was exhibited by the rGOC treated at 190 °C, which was the lowest heat-treatment temperature. The high response comes from the greater amount of oxygen functional groups compared to other rGOC samples, and the tubular structure of the cotton.

2.
Int J Mol Sci ; 23(9)2022 May 09.
Article in English | MEDLINE | ID: mdl-35563670

ABSTRACT

The orphan nuclear receptor 4A1 (NR4A1) is highly expressed in human pancreatic cancer cells and exerts pro-oncogenic activity. In a previous study, we demonstrated that fangchinoline (FCN), a natural inhibitor of nuclear NR4A1, induces NR4A1-dependent apoptosis in human pancreatic cancer cells. In this study, we evaluated FCN and its structural analogs (berbamine, isotetrandrine, tetrandrine, and tubocurarine) for their inhibitory effects on NR4A1 transactivity, and confirmed that tetrandrine (TTD) showed the highest inhibitory effect in pancreatic cancer cells. Moreover, in a tryptophan fluorescence quenching assay, TTD directly bound to the ligand binding domain (LBD) of NR4A1 with a KD value of 10.60 µM. Treatment with TTD decreased proliferation and induced apoptosis in Panc-1 human pancreatic cancer cells in part through the reduced expression of the Sp1-dependent anti-apoptotic gene survivin and induction of ROS-mediated endoplasmic reticulum stress, which are the well-known NR4A1-dependent proapoptotic pathways. Furthermore, at a dose of 25 mg/kg/day, TTD reduced tumor growth in an athymic nude mouse xenograft model bearing Panc-1 cells. These data show that TTD is an NR4A1 antagonist and that modulation of the NR4A1-mediated pro-survival pathways is involved in the antitumor effects of TTD.


Subject(s)
Nuclear Receptor Subfamily 4, Group A, Member 1 , Pancreatic Neoplasms , Animals , Apoptosis , Benzylisoquinolines , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms
3.
Molecules ; 26(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923503

ABSTRACT

The orphan nuclear receptor 4A1 (NR4A1) is overexpressed in pancreatic cancer and exhibits pro-oncogenic activity, and NR4A1 silencing and treatment with its inactivators has been shown to inhibit pancreatic cancer cells and tumor growth. In this study, we identified broussochalcone A (BCA) as a new NR4A1 inhibitor and demonstrated that BCA inhibits cell growth partly by inducing NR4A1-mediated apoptotic pathways in human pancreatic cancer cells. BCA downregulated specificity protein 1 (Sp1)-mediated expression of an anti-apoptotic protein, survivin, and activated the endoplasmic reticulum (ER) stress-mediated apoptotic pathway. These results suggest that NR4A1 inactivation contributes to the anticancer effects of BCA, and that BCA represents a potential anticancer agent targeting NR4A1 that is overexpressed in many types of human cancers.


Subject(s)
Chalcones/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Resorcinols/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Humans , Reactive Oxygen Species/metabolism , Pancreatic Neoplasms
4.
J Biol Chem ; 295(20): 7168-7177, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32241917

ABSTRACT

Serine protease 14 (Prss14)/epithin is a transmembrane serine protease that plays essential roles in tumor progression and metastasis and therefore is a promising target for managing cancer. Prss14/epithin shedding may underlie its activity in cancer and worsen outcomes; accordingly, a detailed understanding of the molecular mechanisms in Prss14/epithin shedding may inform the design of future cancer therapies. On the basis of our previous observation that an activator of PKC, phorbol 12-myristate 13-acetate (PMA), induces Prss14/epithin shedding, here we further investigated the intracellular signaling pathway involved in this process. While using mitogen-activated protein kinase inhibitors to investigate possible effectors of downstream PKC signaling, we unexpectedly found that an inhibitor of c-Jun N-terminal kinase (JNK), SP600125, induces Prss14/epithin shedding even in the absence of PMA. SP600125-induced shedding, like that stimulated by PMA, was mediated by tumor necrosis factor-α-converting enzyme. In contrast, a JNK activator, anisomycin, partially abolished the effects of SP600125 on Prss14/epithin shedding. Moreover, the results from loss-of-function experiments with specific inhibitors, short hairpin RNA-mediated knockdown, and overexpression of dominant-negative PKCßII variants indicated that PKCßII is a major player in JNK inhibition- and PMA-mediated Prss14/epithin shedding. SP600125 increased phosphorylation of PKCßII and tumor necrosis factor-α-converting enzyme and induced their translocation into the plasma membrane. Finally, in vitro cell invasion experiments and bioinformatics analysis of data in The Cancer Genome Atlas breast cancer database revealed that JNK and PKCßII are important for Prss14/epithin-mediated cancer progression. These results provide important information regarding strategies against tumor metastasis.


Subject(s)
Anthracenes/pharmacology , MAP Kinase Kinase 4/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Protein Kinase C beta/metabolism , Protein Kinase Inhibitors/pharmacology , Serine Endopeptidases/metabolism , Signal Transduction/drug effects , Cell Line, Tumor , Humans , MAP Kinase Kinase 4/metabolism , Neoplasm Metastasis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase C beta/genetics , Protein Transport/drug effects , Protein Transport/genetics , Serine Endopeptidases/genetics , Tetradecanoylphorbol Acetate/pharmacology
5.
Int J Obes (Lond) ; 43(5): 952-962, 2019 05.
Article in English | MEDLINE | ID: mdl-30538281

ABSTRACT

BACKGROUND: Suppression of adipogenesis has been considered as a potential target for the prevention and treatment of obesity and associated metabolic disorders, and the nuclear receptor 4A1 (NR4A1/Nur77) and AMPKα are known to play important roles during early and intermediate stages of adipogenesis. Therefore, we hypothesized that dual targeting Nur77 and AMPKα would show strong inhibitory effect on adipogenesis. METHODS: We screened a herbal medicine-based small molecule library to identify novel natural compounds dual targeting Nur77 and AMPKα, and the antiadipogenic effects and mechanisms of action of a "hit" compound were studied in 3T3-L1 cells. In vivo antiobesity effects of the compound were also investigated in high-fat diet (HFD)-induced obese mice. RESULTS: We identified isoalantolactone (ISO) as a new NR4A1 inactivator that also activates AMPKα in 3T3-L1 cells. ISO, as expected, inhibited adipogenic differentiation of 3T3-L1 preadipocytes, accompanied by reduced mitotic clonal expansion (MCE) which occurs in the early stage of adipogenesis and decreased expression of genes required for MCE and cell cycle markers including cyclin A, cyclin D1. Furthermore, ISO reduced body weight gain and fat mass (epididymal, subcutaneous, perirenal, and inguinal white adipose tissues) in the high-fat diet-fed C57BL/6 N mice. Serum levels of triglycerides, aspartate transaminase, and alanine transaminase and hepatic steatosis were also significantly improved in the ISO-treated group compared to the high-fat diet control group. CONCLUSIONS: These results suggest that ISO dual targeting Nur77 and AMPKα during adipogenesis represents a novel class of mechanism-based antiadipogenic agents for treatment of obesity and associated metabolic disorders, including hyperlipidemia and fatty liver.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Polyphenols/pharmacology , Sesquiterpenes/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Diet, High-Fat , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Mice, Obese , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
6.
Acute Crit Care ; 33(4): 271-275, 2018 Nov.
Article in English | MEDLINE | ID: mdl-31723896

ABSTRACT

We experienced a case of severe intraoperative hyperkalemia during laparoscopic radical nephrectomy in a 60-year-old male patient with renal insufficiency, whose hypertension had been managed by preoperative angiotensin II receptor blocker (ARB) and adrenergic beta-antagonist. After renal vessel ligation, his intraoperative potassium concentration suddenly increased to 7.0 mEq/L, but his electrocardiography (ECG) did not show any significant change. While preoperative ARB therapy has been regarded as a contributing factor for further aggravation of underlying renal insufficiency, we assumed that nephrectomy itself and rhabdomyolysis caused by surgical trauma also aggravated the underlying renal dysfunction and resulted in sudden hyperkalemia. Hyperkalemia was managed successfully with calcium gluconate, insulin, furosemide and crystalloid loading during the intraoperative and immediate postoperative periods, and potassium concentration decreased to 5.0 mEq/L at 8 hours after the operation. The patient's hospital course was uncomplicated, but his renal function deteriorated further.

7.
Toxicol Appl Pharmacol ; 332: 32-39, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28754437

ABSTRACT

Previous studies have demonstrated that the orphan nuclear receptor NR4A1 is overexpressed in human pancreatic cancer and antagonizing this receptor promotes apoptosis and inhibits pancreatic cancer cells and tumor growth. In the present study, we identified fangchinoline, a bisbenzyltetrahydroisoquinoline alkaloid from Stephania tetrandra, as a new inactivator of nuclear NR4A1 and demonstrated that fangchinoline inhibits cell proliferation and induces apoptosis, in part, via the NR4A1-dependent pro-apoptotic pathways in human pancreatic cancer cells. It decreased expression of the antiapoptotic protein survivin by inhibiting Sp1-mediated transcription and induced oxidative stress-mediated endoplasmic reticulum (ER) stress in pancreatic cancer cells. These results suggest that inhibition of NR4A1-mediated transcriptional activity was involved in the anticancer effects of fangchinoline, and fangchinoline represents a novel class of mechanism-based anticancer agents targeting NR4A1 that is overexpressed in pancreatic cancer.


Subject(s)
Apoptosis/drug effects , Benzylisoquinolines/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Pancreatic Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum Stress/drug effects , Humans , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Oxidative Stress/drug effects , Pancreatic Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , Stephania tetrandra/chemistry , Survivin
8.
J Anesth ; 31(3): 453-457, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28190124

ABSTRACT

Brachial plexus injury (BPI) often causes severe neuropathic pain that becomes chronic and difficult to treat pharmacologically or surgically. Here, we describe two cases of successful treatment of BPI with peripheral nerve stimulation (PNS). Both patients had experienced severe neuropathic pain after incomplete BPI for a long time (32 and 17 years) and did not response to medication, radiofrequency neuroablation, or spinal cord stimulation. After PNS using ultrasound, their pain was relieved by more than 50% over the course of 1 year. Both patients were satisfied with their improved sleep and quality of life. We conclude that PNS could be an alternative therapeutic modality for neuropathic pain after BPI as it provides direct nerve stimulation, has few complications, and is easy to perform.


Subject(s)
Brachial Plexus Neuropathies/therapy , Electric Stimulation/methods , Neuralgia/therapy , Adult , Brachial Plexus , Humans , Male , Middle Aged , Pain Measurement , Quality of Life , Ultrasonography/methods
9.
Sci Transl Med ; 8(335): 335ra55, 2016 04 20.
Article in English | MEDLINE | ID: mdl-27099174

ABSTRACT

Protection of endothelial integrity has been recognized as a frontline approach to alleviating sepsis progression, yet no effective agent for preserving endothelial integrity is available. Using an unusual anti-angiopoietin 2 (ANG2) antibody, ABTAA (ANG2-binding and TIE2-activating antibody), we show that activation of the endothelial receptor TIE2 protects the vasculature from septic damage and provides survival benefit in three sepsis mouse models. Upon binding to ANG2, ABTAA triggers clustering of ANG2, assembling an ABTAA/ANG2 complex that can subsequently bind and activate TIE2. Compared with a conventional ANG2-blocking antibody, ABTAA was highly effective in augmenting survival from sepsis by strengthening the endothelial glycocalyx, reducing cytokine storms, vascular leakage, and rarefaction, and mitigating organ damage. Together, our data advance the role of TIE2 activation in ameliorating sepsis progression and open a potential therapeutic avenue for sepsis to address the lack of sepsis-specific treatment.


Subject(s)
Antibodies/therapeutic use , Receptor, TIE-2/metabolism , Sepsis/drug therapy , Sepsis/metabolism , Angiopoietin-2/metabolism , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Neutrophil Infiltration/drug effects , Ribonuclease, Pancreatic/metabolism , Vesicular Transport Proteins/metabolism
10.
Biochem Biophys Res Commun ; 452(4): 1084-90, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25245289

ABSTRACT

Epithin/PRSS14, a type II transmembrane serine protease, plays critical roles in cancer metastasis. Previously, we have reported that epithin/PRSS14 undergoes ectodomain shedding in response to phorbol myristate acetate (PMA) stimulation. In this study, we show that transforming growth factor-ß (TGF-ß) induces rapid epithin/PRSS14 shedding through receptor mediated pathway in 427.1.86 thymoma cells. Tumor necrosis factor-α converting enzyme (TACE) is responsible for this shedding. Amino acid sequence encompassing the putative shedding cleavage site of epithin/PRSS14 exhibit strong homology to the cleavage site of l-selectin, a known TACE substrate. TACE inhibitor, TAPI-0 and TACE siRNA greatly reduced TGF-ß-induced epithin/PRSS14 shedding. TGF-ß treatment induces translocation of intracellular pool of TACE to the membrane where epithin/PRSS14 resides. These findings suggest that TGF-ß induces epithin/PRSS14 shedding by mediating translocation of epithin/PRSS14 sheddase, TACE, to the membrane.


Subject(s)
ADAM Proteins/metabolism , Cell Membrane/metabolism , Epithelial Cells/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Transforming Growth Factor beta/metabolism , ADAM17 Protein , Animals , Cell Line , Mice , Protein Transport/physiology
11.
J Biol Chem ; 289(45): 31330-40, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25237190

ABSTRACT

Angiopoietin-2 (Ang-2) not only regulates angiogenesis by binding to its well known receptor Tie2 on endothelial cells but also controls sprouting of Tie2-negative angiogenic endothelial cells and invasion of Tie2-negative non-endothelial cells by binding to integrins. However, the molecular mechanism of the Ang-2/integrin association has been unclear. In this study, we found that the Gln-362 residue of Ang-2 was essential for binding to α5ß1 integrin. A Q362E Ang-2 mutant, which still bound to Tie2, failed to associate with α5ß1 integrin and was unable to activate the integrin downstream signaling of focal adhesion kinase. In addition, unlike wild-type Ang-2, the Q362E Ang-2 mutant was defective in mediating invasion of Tie2-negative glioma or Tie2-positive endothelial cells. Furthermore, the tailpiece domain of the α5 subunit in α5ß1 integrin was critical for binding to Ang-2. Taken together, these results provide a novel insight into the mechanism of integrin regulation by Ang-2, which contributes to tumor invasion and endothelial cell migration in a Tie2-independent manner.


Subject(s)
Angiopoietin-2/metabolism , Endothelial Cells/cytology , Glutamine/metabolism , Integrin alpha5beta1/metabolism , Neoplasms/metabolism , Receptor, TIE-2/metabolism , Animals , CHO Cells , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cricetinae , Cricetulus , Gene Expression Regulation , Humans , Integrins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/pathology , Neovascularization, Pathologic , Plasmids/metabolism , Protein Structure, Tertiary
12.
Bioorg Med Chem ; 20(2): 1139-48, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22196512

ABSTRACT

The well-known 5-HT(1A)/5-HT(7) selectivity issue was tackled by a new series of 4-aminoethylpiperazinyl aryl ketones (1a-1l) specifically designed to distinguish the two hydrophobic sites centered at the anchoring salt bridge. The 4-aminoethylpiperazinyl aryl ketones showed a wide spectrum of activity and selectivity for the 5-HT receptors depending on the type of the hydrophobic groups attached at the aryl piperazinyl ketone scaffold. Docking study of the most active compounds against 5-HT(7)R and 5-HT(1A)R revealed that both receptors have two hydrophobic pockets around the anchoring salt bridge. These two binding sites are perpendicular to each other in 5-HT(7)R but parallel in 5-HT(1A)R, and this observation is well matched with the previous report which claimed that 5-HT(7)R affinity arises from bent conformation of the bound ligand whereas an extended one is best suited for 5-HT(1A)R selectivity. Also, as these pockets have different size and shape, inhibitory activity as well as selectivity of the 4-aminoethylpiperazinyl aryl ketones against 5-HT(7)R and 5-HT(1A)R seemed to be determined by combination of two hydrophobic substituents attached at both ends of the title compounds.


Subject(s)
Ketones/chemistry , Receptor, Serotonin, 5-HT1A/chemistry , Receptors, Serotonin/chemistry , Serotonin Antagonists/chemistry , Binding Sites , Cell Line , Computer Simulation , Drug Design , Humans , Hydrophobic and Hydrophilic Interactions , Ketones/chemical synthesis , Ketones/pharmacology , Piperazines/chemistry , Protein Binding/drug effects , Protein Structure, Tertiary , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Serotonin/metabolism , Serotonin Antagonists/chemical synthesis , Serotonin Antagonists/pharmacology , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 21(10): 3002-6, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21489792

ABSTRACT

A serine-threonine kinase IKK-2 plays an important role in activation of NF-κB through phosphorylation of the inhibitor of NF-κB (IκB). As NF-κB is a major transcription factor that regulates genes responsible for cell proliferation and inflammation, development of selective IKK-2 inhibitors has been an important area of anti-inflammatory and anti-cancer research. In this study, to obtain active and selective IKK-2 inhibitors, various substituents were introduced to a piperidinyl aminopyrimidine core structure. The structure-activity relationship study indicated that hydrogen, methanesulfonyl, and aminosulfonyl groups substituted at the piperidinylamino functionality provide high inhibitory activity against IKK-2. Also, morpholinosulfonyl and piperazinosulfonyl group substituted at the aromatic ring attached to the aminopyrimidine core significantly increased the inhibitory activity of the resulting derivatives. In particular, compound 17 with the aromatic piperazinosulfonyl substituent showed the most potent (IC(50)=1.30 µM) and selective (over other kinases such as p38α, p38ß, JNK1, JNK2, JNK3, and IKK-1) inhibitory activity against IKK-2.


Subject(s)
Drug Discovery , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , I-kappa B Kinase/antagonists & inhibitors , Piperidines/chemistry , Pyrimidines/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Inhibitory Concentration 50 , Molecular Structure , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship
14.
Biochem Biophys Res Commun ; 405(4): 644-50, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21295011

ABSTRACT

PRSS14/Epithin (also known as matriptase and ST14), a member of the type II transmembrane serine proteases, is primarily found in a subpopulation of normal epithelial cells and in epithelial cancers. Its known functions include maintaining the epithelial barrier, thymic development, and cancer progression. In this study, we show that several macrophage cell lines and activated bone marrow-derived macrophages also express PRSS14/Epithin. Surface expression, as well as cytoplasmic expression, was detectable upon activation by IFN-γ, but not TNF-α or TGF-ß. Induction of the protein appeared to be restricted to macrophages. IFN-γ showed a biphasic regulation in RAW264.7 cells, and upregulated expression was sustained for several days. This induction by IFN-γ was partially through the increase of PRSS14/Epithin mRNA production, which is downstream of the JAK pathway, shown by the inhibition by tyrphostin AG490. Using chromatin immunoprecipitation, we verified that two sites among six putative STAT1 binding sites in the PRSS14/Epithin promoter were occupied by STAT1 upon activation. Treatment with IFN-γ enhanced the serum-triggered transendothelial migration of RAW264.7 cells, but not that of PRSS14/Epithin knock-down RAW264.7 cells, although they express multiple markers such as ICAM1, CD80, and CD40 at normal levels. These data strongly suggest that PRSS14/Epithin plays an important role in the transendothelial migration of activated macrophages in the inflammatory microenvironment, and the mode of action is similar to the events in cancer metastasis.


Subject(s)
Cell Movement , Interferon-gamma/metabolism , Janus Kinases/metabolism , Macrophage Activation , Macrophages/immunology , STAT1 Transcription Factor/metabolism , Serine Endopeptidases/metabolism , Animals , Cell Line , Endothelial Cells/immunology , Extracellular Matrix/immunology , Interferon-gamma/pharmacology , Macrophages/drug effects , Membrane Proteins , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , Serine Endopeptidases/genetics
15.
Blood ; 117(4): 1415-24, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21097670

ABSTRACT

Epithin/PRSS14, a type II transmembrane serine protease, is involved in normal epithelial development and tumor progression. Here we report, as an interacting substrate of epithin, a receptor tyrosine kinase Tie2 that is well known for important roles in the vessel stability. Epithin interacts with and degrades the Tie2 extracellular portion that contains the ligand-binding domain. Epithin is located in the neighbor of Tie2-expressing vessels in normal tissue. Furthermore, epithin can cleave and degrade Tie2 not only in the same cell but also from neighboring cells nearby, resulting in the degradation of the Tie2 ectodomain. The remaining Tie2 fragment was highly phosphorylated and was able to recruit a downstream effector, phosphatidylinositol 3-kinase. Knocking down epithin expression using short hairpin RNA in thymoma cell severely impaired the migration through endothelial cells that show the actin rearrangement during the process. The diminution of epithin protein expression in 4T1 breast cancer cells caused the significant decrease in the number of transendothelial migrating cells in vitro as well as in those of metastasizing tumor nodules in vivo, Therefore, we propose that epithin, which regulates endothelial Tie2 functions, plays a critical role in the fine tuning of transendothelial migration for normal and cancer cells.


Subject(s)
Protein Processing, Post-Translational , Receptor, TIE-2/metabolism , Serine Endopeptidases/physiology , Transendothelial and Transepithelial Migration/physiology , Animals , Cells, Cultured , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Humans , Mice , Mice, Inbred BALB C , Models, Biological , Protein Binding/drug effects , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transendothelial and Transepithelial Migration/drug effects , Transendothelial and Transepithelial Migration/genetics , Transfection
16.
Bioorg Med Chem ; 18(21): 7331-7, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20888241

ABSTRACT

Through chelation of the metal ions at the enzyme active site, 1,3-diketoacids (DKAs) show potent inhibition of viral enzymes such as HIV integrase and HCV NS5B. In order to optimize the antiviral activity of the DKAs, structural modification of their metal-binding units, keto-enol acids or monoketo acids, have been actively performed. In this study, we proposed 3-O-arylmethylgalangin 3 as an alternative to ortho-substituted aromatic DKA, a potent inhibitor of HCV NS5B. As a proof-of-concept study, a series of 3-O-arylmethylgalangin derivatives (3a-3r) were prepared and their inhibitory activity against HCV NS5B was estimated. Structure-activity relationship of the 3-O-arylmethylgalangin derivatives was in good accordance with that of the ortho-substituted aromatic DKA series. In particular, two galangin ethers (3g and 3i) completely superimposable with the most potent ortho-substituted aromatic DKA analogue (2) in atom-by-atom fashion showed equipotent inhibitory activity to that of 2. Taken together, this result provides convincing evidence that the 3-O-arylmethylgalangin can successfully mimic the chelating function of the DKA pharmacophore to show potent inhibitory activity against the target enzyme, HCV NS5B.


Subject(s)
Antiviral Agents/chemistry , Flavonoids/chemistry , Hepacivirus/drug effects , Keto Acids/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cell Line, Tumor , Flavonoids/chemical synthesis , Flavonoids/pharmacology , Humans , Keto Acids/chemical synthesis , Keto Acids/pharmacology , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Structure-Activity Relationship , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism
17.
Bioorg Med Chem Lett ; 20(19): 5709-12, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20797857

ABSTRACT

In spite of potent antiviral activity, suboptimal physicochemical properties of aryl diketo acids (ADKs) necessitates modification of the core 1,3-diketo acid functionality into a novel scaffold. As the metal-binding affinity of the diketo acid is the key to the antiviral activity of ADKs, we anticipated 3,5-dihydroxy-4-oxo arrangement of galangin scaffold would serve as an excellent mimic for the diketo acid functionality. In this study, through synthesis and biological evaluation of various galangin derivatives, we have shown that the diketo acid functionality can be successfully replaced with the galangin scaffold by specific combination of the substituents to result in identification of a novel galangin derivative (3s) with anti-HCV activity (EC(50)=0.9 µM) comparable to the ADK counterpart.


Subject(s)
Antiviral Agents/chemistry , Flavonoids/chemistry , Keto Acids/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Flavonoids/chemical synthesis , Flavonoids/pharmacology , Hepacivirus/drug effects , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Structure-Activity Relationship , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism
18.
Biochem Biophys Res Commun ; 395(4): 553-9, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20398629

ABSTRACT

The epithelial-derived type II transmembrane serine protease epithin has been shown to be upregulated in a variety of cancer cell lines and tumor tissues, and its upregulation correlates well with tumor progression in many cases. However, little is known regarding the regulation of its expression and the mechanism of its roles in tumor progression. Here, we show that transforming growth factor-beta (TGF-beta), a potent inducer of epithelial-mesenchymal transition (EMT) in tumor progression, upregulates epithin, and that epithin plays a critical role in TGF-beta-induced EMT. Forced overexpression of epithin induced EMT to exhibit characteristic morphological changes, alternations in EMT-related proteins and enhanced cell motility. Conversely, shRNA-mediated knockdown of endogenous epithin inhibited TGF-beta-induced expression of mesenchymal markers and morphological changes. Furthermore, TGF-beta-induced cell migration and invasion were significantly impaired by epithin knockdown. In addition, we demonstrate that TGF-beta upregulates epithin transcriptionally via the Smad2/Smad4-mediated pathway. These results suggest that epithin is a key mediator of TGF-beta-induced EMT in tumor progression.


Subject(s)
Cell Transdifferentiation/genetics , Epithelium/pathology , Gene Expression Regulation, Neoplastic , Mesoderm/pathology , Neoplasms/pathology , Serine Endopeptidases/genetics , Transforming Growth Factor beta/metabolism , Animals , Dogs , Epithelium/metabolism , Gene Knockdown Techniques , Membrane Proteins , Mesoderm/metabolism , Mice , Neoplasms/genetics , RNA, Small Interfering/genetics , Signal Transduction , Smad2 Protein/metabolism , Smad4 Protein/metabolism , Transcriptional Activation , Up-Regulation
19.
Bioorg Med Chem Lett ; 18(16): 4661-5, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18644717

ABSTRACT

Based on our pharmacophore model of the aryl diketoacids (ADKs), we designed and prepared a series of novel ADK analogues, which showed potent inhibitory activities against the NS5B polymerase in the submicromolar range. Pharmacophore-guided docking study revealed that the antiviral activities of the ADKs are highly dependent upon the aryl linker as well as the size and position of the aromatic substituent. It is of another importance that, unlike previously reported ADKs, three ADK analogues synthesized in this study effectively blocked Hepatitis C Virus (HCV) replication in the replicon systems.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Keto Acids/chemistry , Keto Acids/pharmacology , Viral Nonstructural Proteins/chemistry , Chemistry, Pharmaceutical , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Hepacivirus/metabolism , Inhibitory Concentration 50 , Models, Chemical , Molecular Conformation , Protein Binding , Replicon/drug effects , Viral Nonstructural Proteins/metabolism , Virus Replication
20.
Biosci Biotechnol Biochem ; 67(10): 2083-90, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14586094

ABSTRACT

This study was designed to investigate whether chlorella supplementation may ameliorate oxidative stress and nuclear factor kappa B (NFkappaB) activation in peritoneal macrophages and liver of C57BL/6 mice fed on an atherogenic diet. The animals were maintained on an atherogenic diet (control), or an atherogenic diet supplemented with 3% (w/w) chlorella or 5% (w/w) chlorella for 12 wks. The plasma and hepatic lipid levels were not affected by chlorella supplementation. Hepatic thiobarbituric acid-reactive substances and superoxide anion production in peritoneal macrophages were significantly lower in the 5% chlorella group (p<0.05), but the glutathione level was not altered by chlorella supplementation. The hepatic antioxidative enzyme activities of Cu, Zn-superoxide dismutase and catalase were higher in the mice fed on the 5% chlorella diet (p<0.05). The plasma aspartate aminotransferase activity was lower in the mice fed on the chlorella-containing diets (p<0.05), whereas the alanine aminotransferase activity was not affected by chlorella supplementation. The NFkappaB nuclear binding activities of peritoneal macrophages and liver were significantly lower in the 5% chlorella groups (p<0.05). These results suggest that chlorella supplementation may attenuate oxidative stress by reducing reactive oxygen production and increasing antioxidative processes, thus suppressing inflammatory mediator activation in peritoneal macrophages and liver.


Subject(s)
Chlorella , Liver/metabolism , Macrophages, Peritoneal/metabolism , NF-kappa B/metabolism , Oxidative Stress , Animals , Diet, Atherogenic , Dietary Supplements , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Superoxides/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...