Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 282
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Endocr J ; 71(2): 199-206, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38171884

ABSTRACT

Endometriosis, a common gynecological disorder characterized by the growth of endometrial gland and stroma outside the uterus, causes several symptoms such as dysmenorrhea, hypermenorrhea, and chronic abdominal pain. 17ß estradiol (E2) stimulates the growth of endometriotic lesions. Although estetrol (E4), produced by human fetal liver, is also a natural estrogen, it may have the opposite effects on endometriotic cells. We investigated different effects of E4 and E2 on the invasion and migration of immortalized human endometrial stromal cells (HESCs) and evaluated whether E4 affects the expression of Wiskott-Aldrich syndrome protein (WASP) family member 1 (WASF-1). We measured the invasion of HESCs by a Matrigel chamber assay. Cell migration was measured by wound healing assay and cell tracking analysis. The expression of WASF-1 was confirmed by independent real-time PCR analysis. Transfection of cells with siRNAs was carried out to knock down the expression of WASF-1 in HESCs. E4 significantly inhibited E2-induced invasion and migration of HESCs. WASF-1 was found to be a potential mediator based on metastasis PCR array. WASF-1 was upregulated by E2 and downregulated by E4. Knockdown of WASF-1 inhibited migration. Our results suggest that E4 may inhibit E2-induced growth of endometriotic lesions. Downregulation of WASF-1 is involved in the inhibitory effects of E4 on migration. The use of E4 combined with progestins as combined oral contraceptives may cause endometriotic lesions to regress in women with endometriosis.


Subject(s)
Endometriosis , Estetrol , Humans , Female , Estetrol/metabolism , Estetrol/pharmacology , Endometriosis/metabolism , Endometriosis/pathology , Estrogens/pharmacology , Estradiol/pharmacology , Estradiol/metabolism , Cell Movement , Endometrium/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology
2.
J Nat Prod ; 86(2): 239-245, 2023 02 24.
Article in English | MEDLINE | ID: mdl-36735022

ABSTRACT

Safe and effective nonsteroidal anti-inflammatory drugs are needed. Meanwhile, addition of amino acids to cultures of microorganisms is likely to increase the possibility of novel secondary metabolite isolation. In the course of screening for anti-inflammatory agents using cellular lipopolysaccharide (LPS)-induced nitric oxide (NO) production, two new related compounds with the myceliothermophin structure from a methionine-enriched culture of Myceliophthora thermophila ATCC 42464 were isolated. The new compounds have an additional methylthio group on the myceliothermophin structure and were named myceliostatins A and B. Both compounds inhibited LPS-induced NO production at nontoxic concentrations in macrophage-like mouse monocytic leukemia RAW264.7 cells. Myceliostatin B inhibited the expression of LPS-induced iNOS, IL-6, and IL-1ß and the upstream NF-κB activity in situ and in vitro. Finally, it was found to inhibit NF-κB binding to DNA in the reconstruction system with purified p65. Myceliostatin B also inhibited LPS-induced reactive oxygen species (ROS) production. Thus, myceliostatin B, a novel compound derived from M. thermophila, was found to be a new anti-inflammatory and antioxidant compound directly inhibiting NF-κB.


Subject(s)
Lipopolysaccharides , NF-kappa B , Mice , Animals , NF-kappa B/metabolism , Lipopolysaccharides/pharmacology , Methionine , Anti-Inflammatory Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Nitric Oxide/metabolism
3.
Childs Nerv Syst ; 39(6): 1519-1528, 2023 06.
Article in English | MEDLINE | ID: mdl-36807999

ABSTRACT

PURPOSE: While pediatric glioblastomas are molecularly distinct from adult counterparts, the activation of NF-kB is partially shared by both subsets, playing key roles in tumor propagation and treatment response. RESULTS: We show that, in vitro, dehydroxymethylepoxyquinomicin (DHMEQ) impairs growth and invasiveness. Xenograft response to the drug alone varied according to the model, being more effective in KNS42-derived tumors. In combination, SF188-derived tumors were more sensitive to temozolomide while KNS42-derived tumors responded better to the combination with radiotherapy, with continued tumor regression. CONCLUSION: Taken together, our results strengthen the potential usefulness of NF-kB inhibition in future therapeutic strategies to overcome this incurable disease.


Subject(s)
Glioblastoma , NF-kappa B , Child , Humans , NF-kappa B/metabolism , NF-kappa B/pharmacology , Glioblastoma/drug therapy , Apoptosis , Cell Line, Tumor
4.
Hepatology ; 74(1): 83-98, 2021 07.
Article in English | MEDLINE | ID: mdl-33434356

ABSTRACT

BACKGROUND AND AIMS: Chronic HBV infection is a major health problem worldwide. Currently, the first-line treatment for HBV is nucleos(t)ide analogs or interferons; however, efficient therapeutic approaches that enable cure are lacking. Therefore, anti-HBV agents with mechanisms distinct from those of current drugs are needed. Sodium taurocholate cotransporting polypeptide (NTCP) was previously identified as an HBV receptor that is inhibited by several compounds. Farnesoid X receptor (FXR) activation also inhibits NTCP function. APPROACH AND RESULTS: In this study, we investigated the inhibitory effect of bile acid (BA) derivatives-namely obeticholic acid (OCA), 6α-ethyl-24-nor-5ß-cholane-3α,7α,23-triol-23 sulfate sodium salt (INT-767; a dual agonist of FXR and Takeda G protein-coupled receptor [TGR5]), and 6α-ethyl-23(S)-methyl-cholic acid (INT-777; a TGR5 agonist)-3-(2,6-dichlorophenyl)-4-(3'-carboxy-2-chlorostilben-4-yl)oxymethyl-5-isopropylisoxazole (GW4064; a FXR agonist), cyclosporin A, and irbesartan. OCA and INT-777 suppressed HBV infection in HepG2-human NTCP-C4 cells. Interestingly, INT-767 showed potent inhibition by attaching to HBV particles rather than binding to NTCP. As an entry inhibitor, INT-767 was stronger than various natural BAs. Furthermore, in chimeric mice with humanized liver, INT-767 markedly delayed the initial rise of HBsAg, HBeAg, and HBV DNA and reduced covalently closed circular DNA. The strong inhibitory effect of INT-767 may be due to the cumulative effect of its ability to inhibit the entry of HBV and to stimulate FXR downstream signaling, which affects the postentry step. CONCLUSIONS: Our results suggest that BA derivatives, particularly INT-767, are prospective candidate anti-HBV agents. Clarifying the underlying mechanisms of BA derivatives would facilitate the development of anti-HBV agents.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B, Chronic/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, G-Protein-Coupled/agonists , Virus Internalization/drug effects , Animals , Antiviral Agents/therapeutic use , Bile Acids and Salts/pharmacology , Bile Acids and Salts/therapeutic use , Chenodeoxycholic Acid/analogs & derivatives , Chenodeoxycholic Acid/pharmacology , Chenodeoxycholic Acid/therapeutic use , Cholic Acids/pharmacology , Cholic Acids/therapeutic use , Disease Models, Animal , Hep G2 Cells , Hepatitis B virus/drug effects , Hepatitis B virus/metabolism , Hepatitis B, Chronic/virology , Humans , Male , Mice , Organic Anion Transporters, Sodium-Dependent/metabolism , Prospective Studies , Receptors, Cytoplasmic and Nuclear/metabolism , Symporters/metabolism , Transplantation Chimera
5.
J Clin Lab Anal ; 36(1): e24146, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34837712

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19) and is capable of human-to-human transmission and rapid global spread. Thus, the establishment of high-quality viral detection and quantification methods, and the development of anti-SARS-CoV-2 agents are critical. METHODS: Here, we present the rapid detection of infectious SARS-CoV-2 particles using a plaque assay with 0.5% agarose-ME (Medium Electroosmosis) as an overlay medium. RESULTS: The plaques were capable of detecting the virus within 36-40 h post-infection. In addition, we showed that a monogalactosyl diacylglyceride isolated from a microalga (Coccomyxa sp. KJ) could inactivate the clinical isolates of SARS-CoV-2 in a time- and concentration-dependent manner. CONCLUSIONS: These results would allow rapid quantification of the infectious virus titers and help develop more potent virucidal agents against SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Galactose/analogs & derivatives , Glycerides/pharmacology , Microalgae/chemistry , SARS-CoV-2/drug effects , Animals , Antiviral Agents/chemistry , COVID-19/virology , Chlorocebus aethiops , Chlorophyta/chemistry , Galactose/chemistry , Galactose/pharmacology , Glycerides/chemistry , Humans , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Vero Cells , Viral Plaque Assay
6.
Chem Pharm Bull (Tokyo) ; 70(7): 477-482, 2022.
Article in English | MEDLINE | ID: mdl-35786566

ABSTRACT

1,2-Naphthoquinone (2-NQ) is a nucleophile acceptor that non-selectively makes covalent bonds with cysteine residues in various cellular proteins, and is also found in diesel exhaust, an air pollutant. This molecule has rarely been considered as a pharmacophore of bioactive compounds, in contrast to 1,4-naphthoquinone. We herein designed and synthesized a compound named N-(7,8-dioxo-7,8-dihydronaphthalen-1-yl)-2-methoxybenzamide (MBNQ), in which 2-NQ was hybridized with the nuclear factor-κB (NF-κB) inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) as a nucleophile acceptor. Although 50 µM MBNQ did not inhibit NF-κB signaling, 10 µM MBNQ induced cell death in the lung cancer cell line A549, which was insensitive to 2-NQ (10 µM). In contrast, MBNQ was less toxic in normal lung cells than 2-NQ. A mechanistic study showed that MBNQ mainly induced apoptosis, presumably via the activation of p38 mitogen-activated protein kinase (MAPK). Collectively, the present results demonstrate that the introduction of an appropriate substituent into 2-NQ constitutes a new biologically active entity, which will lead to the development of 2-NQ-based drugs.


Subject(s)
Lung Neoplasms , Naphthoquinones , Apoptosis , Humans , Lung Neoplasms/drug therapy , NF-kappa B/metabolism , Naphthoquinones/pharmacology
7.
ScientificWorldJournal ; 2022: 8454865, 2022.
Article in English | MEDLINE | ID: mdl-36330350

ABSTRACT

Background: Hot water extract of Sasa albomarginata (Kumazasa) leaves is commercially available and used as a dietary supplement or skincare cream. The extract possesses anti-inflammatory activity on the mouse atopic dermatitis model. To elucidate the mechanism of in vivo activity, we have studied the cellular anti-inflammatory and antioxidant activities of the extract and its constituents. Methods: Secretion of mouse and human IL-6 was measured by ELISA. ROS production was measured by a fluorescent reagent. Ultrahigh performance liquid chromatography (UHPLC)/MS was used for the ingredient analysis. Results: The Sasa albomarginata extract inhibited inflammatory mediators such as LPS-induced NO, IL-6, and ROS production in mouse monocyte leukemia RAW264.7 cells. It also inhibited iNOS, IL-6, and IL-1ß expressions. Moreover, it inhibited LPS-induced IL-6 expression and production in human monocyte leukemia THP-1 cells differentiated into macrophages. The HPLC analysis of the extract revealed the existence of coumaric acid, ferulic acid, and coumaric acid methyl ester. Coumaric acid methyl ester but not coumaric acid or ferulic acid inhibited LPS-induced NO, IL-6, and ROS production in RAW264.7 cells and IL-6 production in differentiated THP-1 cells. Conclusion: The hot water extract of Sasa albomarginata leaves and one of its constituents possess cellular anti-inflammatory and antioxidant activities.


Subject(s)
Leukemia , Sasa , Humans , Antioxidants/pharmacology , Lipopolysaccharides , Interleukin-6 , Esters , Reactive Oxygen Species , Plant Extracts/pharmacology , Plant Extracts/chemistry , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Water , Nitric Oxide/metabolism
8.
Mar Drugs ; 19(4)2021 Apr 05.
Article in English | MEDLINE | ID: mdl-33916424

ABSTRACT

Not only physiological phenomena but also pathological phenomena can now be explained by the change of signal transduction in the cells of specific tissues. Commonly used cellular signal transductions are limited. They consist of the protein-tyrosine kinase dependent or independent Ras-ERK pathway, and the PI3K-Akt, JAK-STAT, SMAD, and NF-κB-activation pathways. In addition, biodegradation systems, such as the ubiquitin-proteasome pathway and autophagy, are also important for physiological and pathological conditions. If we can control signaling for each by a low-molecular-weight agent, it would be possible to treat diseases in new ways. At present, such cell signaling inhibitors are mainly looked for in plants, soil microorganisms, and the chemical library. The screening of bioactive metabolites from deep-sea organisms should be valuable because of the high incidence of finding novel compounds. Although it is still an emerging field, there are many successful examples, with new cell signaling inhibitors. In this review, we would like to explain the current view of the cell signaling systems important in diseases, and show the inhibitors found from deep-sea organisms, with their structures and biological activities. These inhibitors are possible candidates for anti-inflammatory agents, modulators of metabolic syndromes, antimicrobial agents, and anticancer agents.


Subject(s)
Anti-Infective Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents/pharmacology , Aquatic Organisms/metabolism , Signal Transduction/drug effects , Animals , Anti-Infective Agents/isolation & purification , Anti-Inflammatory Agents/isolation & purification , Antineoplastic Agents/isolation & purification , Humans , Molecular Structure , Secondary Metabolism , Structure-Activity Relationship
9.
Mar Drugs ; 18(12)2020 Nov 26.
Article in English | MEDLINE | ID: mdl-33256194

ABSTRACT

In the present study, four new compounds including a pair of 2-benzoyl tetrahydrofuran enantiomers, namely, (-)-1S-myrothecol (1a) and (+)-1R-myrothecol (1b), a methoxy-myrothecol racemate (2), and an azaphilone derivative, myrothin (3), were isolated along with four known compounds (4-7) from cultures of the deep-sea fungus Myrothecium sp. BZO-L062. Enantiomeric compounds 1a and 1b were separated through normal-phase chiral high-performance liquid chromatography. The absolute configurations of 1a, 1b, and 3 were assigned by ECD spectra. Among them, the new compound 1a and its enantiomer 1b exhibited anti-inflammatory activity, inhibited nitric oxide formation in lipopolysaccharide-treated RAW264.7 cells, and exhibited antioxidant activity in the 2,2-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) and oxygen radical absorbance capacity assays.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Hypocreales/metabolism , Macrophages/drug effects , Animals , Anti-Inflammatory Agents/isolation & purification , Antioxidants/isolation & purification , Geologic Sediments/microbiology , Inflammation Mediators/metabolism , Macrophages/metabolism , Mice , Molecular Structure , Nitric Oxide/metabolism , Oxygen Radical Absorbance Capacity , RAW 264.7 Cells , Structure-Activity Relationship
10.
J Biol Chem ; 293(52): 20214-20226, 2018 12 28.
Article in English | MEDLINE | ID: mdl-30377255

ABSTRACT

Conophylline is a Vinca alkaloid from leaves of the tropical plant Ervatamia microphylla and has been shown to mimic the effect of the growth and differentiation factor activin A on pancreatic progenitor cells. However, activin A stimulates fibrosis of pancreatic stellate cells, whereas conophylline inhibits it, suggesting that this compound may serve as an antifibrotic drug. Here we investigated the effects of conophylline on human foreskin fibroblasts, especially focusing on extracellular matrix (ECM) proteins. A gene microarray analysis revealed that conophylline remarkably suppressed expression of the gene for hyaluronan synthase 2 (HAS2) and of its antisense RNA, whereas the expression of collagen genes was unaffected. Of note, immunostaining experiments revealed that conophylline substantially inhibits incorporation of versican and collagens into the ECM in cells treated with transforming growth factor ß (TGFß), which promotes collagen synthesis, but not in cells not treated with TGFß. Moreover, a protein biosynthesis assay disclosed that conophylline decreases collagen biosynthesis, concomitant with a decrease in total protein biosynthesis, indicating that conophylline-mediated inhibition of fibrosis is not specific to collagen synthesis. Conophylline affected neither TGFß-induced nuclear translocation of SMAD family member 2/3 (SMAD2/3) nor phosphorylation of SMAD2. However, conophylline substantially inhibited phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2), suggesting that conophylline inhibits HAS2 expression via TGFß-mediated activation of the ERK1/2 pathway. Taken together, our results indicate that conophylline may be a useful inhibitor of ECM formation in fibrosis.


Subject(s)
Extracellular Matrix/metabolism , MAP Kinase Signaling System/drug effects , Vinca Alkaloids/pharmacology , Cells, Cultured , Collagen/metabolism , Fibroblasts , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hyaluronan Synthases/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Protein Biosynthesis/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Versicans/metabolism
11.
Cancer Sci ; 110(1): 334-344, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30353606

ABSTRACT

Despite recent advances in cancer treatment, pancreatic cancer is a highly malignant tumor type with a dismal prognosis and it is characterized by dense desmoplasia in the cancer tissue. Cancer-associated fibroblasts (CAF) are responsible for this fibrotic stroma and promote cancer progression. We previously reported that a novel natural compound conophylline (CnP) extracted from the leaves of a tropical plant reduced liver and pancreatic fibrosis by suppression of stellate cells. However, there have been no studies to investigate the effects of CnP on CAF, which is the aim of this work. Here, we showed that CAF stimulated indicators of pancreatic cancer malignancy, such as proliferation, invasiveness, and chemoresistance. We also showed that CnP suppressed CAF activity and proliferation, and inhibited the stimulating effects of CAF on pancreatic cancer cells. Moreover, CnP strongly decreased the various cytokines involved in cancer progression, such as interleukin (IL)-6, IL-8, C-C motif chemokine ligand 2 (CCL2), and C-X-C motif chemokine ligand 12 (CXCL12), secreted by CAF. In vivo, CAF promoted tumor proliferation and desmoplastic formation in a mouse xenograft model, CnP reduced desmoplasia of tumors composed of pancreatic cancer cells + CAF, and combination therapy of CnP with gemcitabine remarkably inhibited tumor proliferation. Our findings suggest that CnP is a promising therapeutic strategy of combination therapy with anticancer drugs to overcome refractory pancreatic cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cytokines/metabolism , Pancreatic Neoplasms/prevention & control , Xenograft Model Antitumor Assays , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cytokines/genetics , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Inbred NOD , Mice, SCID , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics , Vinca Alkaloids/administration & dosage , Gemcitabine
12.
Immunopharmacol Immunotoxicol ; 41(1): 32-39, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30724631

ABSTRACT

Background: Dehydroxymethylepoxyquinomicin (DHMEQ) is a specific and potent inhibitor of nuclear factor-kappa B (NF-κB) and has been shown to possess promising potential as an anti-inflammation including anti-atopic dermatitis (AD)-like skin lesions. Objective: To further evaluate the activity of DHMEQ in vivo modified AD-like lesion model in BALB/c mice and in vitro AD-like lesion cell model in human keratinocytes. Materials and methods: In this study, in vivo modified AD-like lesion model in BALB/c mice was chronically induced by the repetitive and alternative application of 2,4-dinitrochlorobenzene (DNCB) and oxazolone (OX) on ears, and stratum corneum of the ear skin was additionally stripped off with surgical tapes before each challenge with DNCB/OX. Moreover, in vitro AD-like lesion cell model in human keratinocytes (HaCaT) achieved by stimulating HaCaT cells with tumor necrosis factor (TNF)-α plus interferon (IFN)-γ was used to investigate mechanisms of the action. Results: The lesions derived from the stratum corneum-removed AD-like lesion model reaches to peak as well as DHMEQ arrives to its efficacy a week earlier than the data previously obtained from the common AD-like lesion model. Results showed that the drug reduced the ear thickness, epidermal thickness, mast cell infiltration, and gene expressions of interleukin (IL)-4, IL-13, and interferon (IFN)-γ in ear tissues. It significantly inhibited the expression of cytokines IL-6 and IL-1ß, chemokines thymus and activation-regulated chemokine (TARC)/CCL17, and macrophage-derived chemokine (MDC)/CCL22 in the stimulated HaCaT cells. Discussion and conclusion: This study indicated that the action of DHMEQ's anti-AD like lesions might be related to its inhibition on NF-κB.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Benzamides/therapeutic use , Cyclohexanones/therapeutic use , Dermatitis, Atopic/prevention & control , Epidermis/drug effects , NF-kappa B/antagonists & inhibitors , Animals , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/immunology , Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Dinitrochlorobenzene , Disease Models, Animal , Epidermis/immunology , Epidermis/pathology , Female , Humans , Keratinocytes/drug effects , Keratinocytes/immunology , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/immunology
14.
Haematologica ; 103(1): 136-147, 2018 01.
Article in English | MEDLINE | ID: mdl-29122993

ABSTRACT

Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) is known to play an important role in the pathogenesis of chronic lymphocytic leukemia (CLL). Several NF-κB inhibitors were shown to successfully induce apoptosis of CLL cells in vitro Since the microenvironment is known to be crucial for the survival of CLL cells, herein, we tested whether NF-κB inhibition may still induce apoptosis in these leukemic cells in the presence of protective stromal interaction. We used the specific NF-κB inhibitor dehydroxymethylepoxyquinomicin (DHMEQ). Microenvironmental support was mimicked by co-culturing CLL cells with bone marrow-derived stromal cell lines (HS-5 and M2-10B4). NF-κB inhibition by DHMEQ in CLL cells could be confirmed in both the monoculture and co-culture setting. In line with previous reports, NF-κB inhibition induced apoptosis in the monoculture setting by activating the intrinsic apoptotic pathway resulting in poly (ADP-ribose) polymerase (PARP)-cleavage; however, it was unable to induce apoptosis in leukemic cells co-cultured with stromal cells. Similarly, small interfering ribonucleic acid (siRNA)-mediated RELA downregulation induced apoptosis of CLL cells cultured alone, but not in the presence of supportive stromal cells. B-cell activating factor (BAFF) was identified as a microenvironmental messenger potentially protecting the leukemic cells from NF-κB inhibition-induced apoptosis. Finally, we show improved sensitivity of stroma-supported CLL cells to NF-κB inhibition when combining the NF-κB inhibitor with the SYK inhibitor R406 or the Bruton's tyrosine kinase (BTK) inhibitor ibrutinib, agents known to inhibit the stroma-leukemia crosstalk. We conclude that NF-κB inhibitors are not promising as monotherapies in CLL, but may represent attractive therapeutic partners for ibrutinib and R406.


Subject(s)
Apoptosis/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mesenchymal Stem Cells/metabolism , NF-kappa B/antagonists & inhibitors , Tumor Microenvironment , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Biomarkers , Cell Line, Tumor , Cell Survival/drug effects , Coculture Techniques , Cyclohexanones/pharmacology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
15.
Parasitology ; 145(14): 1890-1895, 2018 12.
Article in English | MEDLINE | ID: mdl-29739480

ABSTRACT

We have previously shown that the C-terminal region of the intermediate subunit of Entamoeba histolytica galactose- and N-acetyl-D-galactosamine-inhibitable lectin (C-Igl) is a useful antigen for serodiagnosis of amebiasis. An immunochromatographic kit was developed using fluorescent silica nanoparticles coated with C-Igl prepared in Escherichia coli. Samples for examination were added to the freeze-dried particles and then applied to the immunochromatographic device, in which a test line on the membrane was also coated with C-Igl. Fluorescent intensity was measured using a hand-held reader. In an evaluation of the kit using a human monoclonal antibody, the minimum amount of C-Igl specific antibody showing positive results was 100 pg. In the evaluation of serum samples with different antibody titers in indirect immunofluorescent antibody tests in the kit, 20 µL of serum was sufficient to obtain positive results at 30 min. Serum samples from symptomatic patients with amebic colitis and amebic liver abscess and those from asymptomatic E. histolytica-cyst carriers showed positive results in the kit. Based on evaluation using sera from healthy controls and patients with other infectious diseases, the sensitivity and specificity of the kit were 100 and 97.6%, respectively. Therefore, we conclude that the newly developed kit is useful for rapid serodiagnosis of amebiasis.


Subject(s)
Amebiasis/diagnosis , Antibodies, Protozoan/blood , Chromatography, Affinity/instrumentation , Reagent Kits, Diagnostic , Serologic Tests/instrumentation , Antibodies, Monoclonal/immunology , Antigens, Protozoan/blood , Dysentery, Amebic/diagnosis , Entamoeba histolytica , Entamoebiasis/diagnosis , Humans , Liver Abscess, Amebic/diagnosis , Nanoparticles , Sensitivity and Specificity , Silicon Dioxide
16.
Int J Mol Sci ; 19(3)2018 Mar 03.
Article in English | MEDLINE | ID: mdl-29510517

ABSTRACT

We previously designed and synthesized dehydroxyepoxyquinomicin (DHMEQ) as an inhibitor of NF-κB based on the structure of microbial secondary metabolite epoxyquinomicin C. DHMEQ showed anti-inflammatory and anticancer activity in various in vivo disease models without toxicity. On the other hand, the process of cancer metastasis consists of cell detachment from the primary tumor, invasion, transportation by blood or lymphatic vessels, invasion, attachment, and formation of secondary tumor. Cell detachment from the primary tumor and subsequent invasion are considered to be early phases of metastasis, while tumor cell attachment to the tissue and secondary tumor formation the late phases. The assay system for the latter phase was set up with intra-portal-vein injection of pancreatic cancer cells. Intraperitoneal administration of DHMEQ was found to inhibit liver metastasis possibly by decreasing the expression of MMP-9 and IL-8. Also, when the pancreatic cancer cells treated with DHMEQ were inoculated into the peritoneal cavity of mice, the metastatic foci formation was inhibited. These results indicate that DHMEQ is likely to inhibit the late phase of metastasis. Meanwhile, we have recently employed three-dimensional (3D) culture of breast cancer cells for the model of early phase metastasis, since the 3D invasion just includes cell detachment and invasion into the matrix. DHMEQ inhibited the 3D invasion of breast cancer cells at 3D-nontoxic concentrations. In this way, DHMEQ was shown to inhibit the late and early phases of metastasis. Thus, DHMEQ is likely to be useful for the suppression of cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cyclohexanones/pharmacology , NF-kappa B/antagonists & inhibitors , Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Benzamides/chemistry , Benzamides/therapeutic use , Cyclohexanones/chemistry , Cyclohexanones/therapeutic use , Humans , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Quinones/chemistry , Quinones/pharmacology
17.
J Biol Chem ; 291(14): 7373-85, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26839315

ABSTRACT

Choroidal neovascularization (CNV) is a pathogenic process of age-related macular degeneration, a vision-threatening disease. The retinal pigment epithelium and macrophages both influence CNV development. However, the underlying mechanisms remain obscure. Here, we focus on Angptl2 (angiopoietin-like protein 2), a cytokine involved in age-related systemic diseases. Angptl2 was originally identified as an adipocytokine and is also expressed in the eye. Using a laser-induced CNV model, we found thatAngptl2KO mice exhibited suppressed CNV development with reduced macrophage recruitment and inflammatory mediator induction. The mediators monocyte chemotactic protein-1, interleukin-1ß (Il-1ß),Il-6, matrix metalloprotease-9 (Mmp-9), and transforming growth factor-ß1 (Tgf-ß1) that were up-regulated during CNV development were all suppressed in the retinal pigment epithelium-choroid of CNV models generated in theAngptl2KO mice. Bone marrow transplantation using wild-type and KO mice suggested that both bone marrow-derived and host-derived Angptl2 were responsible for macrophage recruitment and CNV development. Peritoneal macrophages derived fromAngptl2KO mice expressed lower levels of the inflammatory mediators. In the wild-type peritoneal macrophages and RAW264.7 cells, Angptl2 induced the mediators via integrins α4 and ß2, followed by the downstream activation of NF-κB and ERK. The activation of NF-κB and ERK by Angptl2 also promoted macrophage migration. Therefore, Angptl2 from focal tissue might trigger macrophage recruitment, and that from recruited macrophages might promote expression of inflammatory mediators including Angptl2 in an autocrine and/or paracrine fashion to facilitate CNV development. Angptl2 might therefore represent a multistep regulator of CNV pathogenesis and serve as a new therapeutic target for age-related macular degeneration.


Subject(s)
Angiopoietins/biosynthesis , Choroidal Neovascularization/metabolism , Macrophages/metabolism , Macular Degeneration/metabolism , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/genetics , Animals , CD18 Antigens/genetics , CD18 Antigens/metabolism , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Choroidal Neovascularization/genetics , Choroidal Neovascularization/pathology , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Integrin alpha4/genetics , Integrin alpha4/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/pathology , Macular Degeneration/genetics , Macular Degeneration/pathology , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
18.
Biochem Biophys Res Commun ; 485(1): 76-81, 2017 03 25.
Article in English | MEDLINE | ID: mdl-28188787

ABSTRACT

The three-dimensional (3D) culture of cancer cells provides an environmental condition closely related to the condition in vivo. It would especially be an ideal model for the early phase of metastasis, including the detachment and invasion of cancer cells from the primary tumor. In one hand, dehydroxymethylepoxyquinomicin (DHMEQ), an NF-κB inhibitor, is known to inhibit cancer progression and late phase metastasis in animal experiments. In the present research, we studied the inhibitory activity on the 3D invasion of breast carcinoma cells. Breast carcinoma MDA-MB-231 cells showed the most active invasion from spheroid among the cell lines tested. DHMEQ inhibited the 3D invasion of cells at the 3D-nontoxic concentrations. The PCR array analysis using RNA isolated from the 3D on-top cultured cells indicated that matrix metalloproteinase (MMP)-2 expression is lowered by DHMEQ. Knockdown of MMP-2 and an MMP inhibitor, GM6001, both inhibited the invasion. DHMEQ was shown to inhibit the promoter activity of MMP-2 in the reporter assay. Thus, DHMEQ was shown to inhibit NF-κB/MMP-2-dependent cellular invasion in 3D-cultured MDA-MB-231 cells, suggesting that DHMEQ would inhibit the early phase of metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Breast Neoplasms/drug therapy , Cyclohexanones/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase Inhibitors/pharmacology , NF-kappa B/antagonists & inhibitors , Neoplasm Invasiveness/prevention & control , Breast/drug effects , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Promoter Regions, Genetic/drug effects
19.
Bioorg Med Chem Lett ; 27(3): 562-566, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28003138

ABSTRACT

(-)-Dehydroxymethylepoxyquinomicin ((-)-DHMEQ, 1) is a specific inhibitor of NF-κB. It binds to SH group in the specific cysteine residue of NF-κB components with its epoxide moiety to inhibit DNA binding. In the present research, we have designed and synthesized an epoxide-free analog called (S)-ß-salicyloylamino-α-exo-methylene-Æ´-butyrolactone (SEMBL, 3). SEMBL inhibited DNA binding of NF-κB component p65 in vitro. It inhibited LPS-induced NF-κB activation, iNOS expression, and inflammatory cytokine secretions. It also inhibited NF-κB and cellular invasion in ovarian carcinoma ES-2 cells. Moreover, its stability in aqueous solution was greatly enhanced compared with (-)-DHMEQ. Thus, SEMBL has a potential to be a candidate for a new anti-inflammatory and anticancer agent.


Subject(s)
4-Butyrolactone/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Drug Design , NF-kappa B/antagonists & inhibitors , Salicylamides/pharmacology , 4-Butyrolactone/chemical synthesis , 4-Butyrolactone/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Mice , Molecular Structure , NF-kappa B/metabolism , RAW 264.7 Cells , Salicylamides/chemical synthesis , Salicylamides/chemistry , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 27(16): 3862-3866, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28666734

ABSTRACT

Protein prenylation such as farnesylation and geranylgeranylation is associated with various diseases. Thus, many inhibitors of prenyltransferase have been developed. We report novel inhibitors of farnesyltransferase with a zinc-site recognition moiety and a farnesyl/dodecyl group. Molecular docking analysis showed that both parts of the inhibitor fit well into the catalytic domain of farnesyltransferase. The synthesized inhibitors showed activity against farnesyltransferase in vitro and inhibited proliferation of the pancreatic cell line AsPC-1. Among the compounds with farnesyl and dodecyl groups, the inhibitor with a farnesyl group was found to have stronger and more selective activity.


Subject(s)
Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/antagonists & inhibitors , Organometallic Compounds/pharmacology , Zinc/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Farnesyltranstransferase/metabolism , Humans , Molecular Docking Simulation , Molecular Structure , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Structure-Activity Relationship , Zinc/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL