Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Invest New Drugs ; 39(1): 111-121, 2021 02.
Article in English | MEDLINE | ID: mdl-32885355

ABSTRACT

Primary effusion lymphoma (PEL), caused by Kaposi's sarcoma-associated herpesvirus (KSHV), presents as a lymphomatous effusion in body cavities and has a poor prognosis. The anti-malaria drug, artesunate, possesses anti-neoplastic potential. Therefore, we aimed to investigate its effect on KSHV-infected PEL cell lines. Artesunate inhibited cell growth and viability of PEL cells, but its effect on peripheral blood mononuclear cells was less pronounced. Artesunate induced G1 phase arrest by downregulating cyclin D1/D2, CDK2/6 and c-Myc. Artesunate increased reactive oxygen species and DNA damage, but did not affect the expression of latent and lytic genes of KSHV. It exhibited cytotoxicity through caspase-dependent and -independent pathways and reduced Bcl-xL, survivin, XIAP and c-IAP1/2 levels. Furthermore, artesunate suppressed NF-κB and AP-1 by inhibiting IκB kinase and IκBα phosphorylation as well as JunB expression. Finally, artesunate treatment attenuated PEL development in mice. Our data support that artesunate is a potential drug for PEL treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Artesunate/pharmacology , Herpesvirus 8, Human/drug effects , Lymphoma, Primary Effusion/pathology , Animals , Apoptosis/drug effects , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Damage/drug effects , Female , Herpesvirus 8, Human/genetics , Humans , I-kappa B Kinase/drug effects , Mice , Mice, SCID , NF-KappaB Inhibitor alpha/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
2.
ACS Infect Dis ; 5(10): 1764-1771, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31434474

ABSTRACT

Kinase inhibitors comprise a diverse cohort of chemical scaffolds that are active in multiple biological systems. Currently, thousands of eukaryotic kinase inhibitors are commercially available, have well-characterized targets, and often carry pharmaceutically favorable toxicity profiles. Recently, our group disclosed that derivatives of the natural product meridianin D, a known inhibitor of eukaryotic kinases, modulated behaviors of both Gram-positive and Gram-negative bacteria. Herein, we expand our exploration of kinase inhibitors in Gram-negative bacilli utilizing three commercially available kinase inhibitor libraries and, ultimately, identify two chemical structures that potentiate colistin (polymyxin E) in multiple strains. We report IMD-0354, an inhibitor of IKK-ß, as a markedly effective adjuvant in colistin-resistant bacteria and also describe AR-12 (OSU-03012), an inhibitor of pyruvate dehydrogenase kinase-1 (PDK-1), as a potentiator in colistin-sensitive strains. This report comprises the first description of the novel cross-reactivity of these molecules.


Subject(s)
Adjuvants, Pharmaceutic/pharmacology , Colistin/pharmacology , Gram-Negative Bacteria/drug effects , Protein Kinase Inhibitors/pharmacology , Adjuvants, Pharmaceutic/chemistry , Benzamides/pharmacology , Cell Line , Colistin/chemistry , Drug Combinations , Drug Resistance, Multiple, Bacterial/drug effects , Eukaryota , Gram-Positive Bacteria/drug effects , I-kappa B Kinase/drug effects , Lipid A , Microbial Sensitivity Tests , Pyrazoles/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/drug effects , Sulfonamides/pharmacology
3.
Mol Pharm ; 15(11): 5244-5251, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30265541

ABSTRACT

Aseptic loosening of artificial joints mainly accounts for the failure of arthroplasty. We previously reported that ursolic acid (UA) inhibited osteolysis caused by titanium (Ti) wear particles via suppression of NF-kB signaling. In the present study, that the suppressive effect of UA on Ti-particle-induced inflammation and osteoclastogenesis targets on IKKß cys-179 was demonstrated. A retrovirus packaged IKKßC179A plasmid with a Cys-179 mutation replaced by Ala was constructed. qRT-PCR, immunoblot, and immunofluorescence were used to evaluate the gene expressions. Secreted inflammatory cytokines were detected by ELISA. Formation and function of osteoclastogenesis were evaluated by TRAP stain and hydroxylapatite resorption assays. As a result, a mutation of IKKßC179A rescued the therapeutic effect of UA on Ti-particle-induced inflammation, including morphological transforms, upregulation of iNOS and COX-2, increased secretions of TNF-α, IL-1ß, and IL-6, and decreased secretion of IL-10. Meanwhile, inhibition of osteoclastogenesis and hydroxylapatite resorptions were restored by transfection of IKKßC179A. Phosphorylations of p65 and the IKKα/ß complex and translocation of p65 into the nucleus were suppressed by UA but rescued by a mutation of IKKßC179A. Conclusively, UA inhibits Ti-wear-particle-induced inflammation, osteoclastogenesis, and hydroxylapatite resorption via modifying cysteine 179 of IKKß.


Subject(s)
Bone Resorption/prevention & control , I-kappa B Kinase/drug effects , Inflammation/prevention & control , Titanium/adverse effects , Triterpenes/pharmacology , Animals , Bone Resorption/chemically induced , Bone Resorption/pathology , Cysteine/chemistry , Cysteine/drug effects , Durapatite/metabolism , I-kappa B Kinase/chemistry , I-kappa B Kinase/genetics , Inflammation/chemically induced , Inflammation/pathology , Joint Prosthesis/adverse effects , Mice , Mice, Inbred C57BL , Monocytes , Osteoclasts/drug effects , Osteoclasts/metabolism , Primary Cell Culture , Prosthesis Failure/drug effects , RAW 264.7 Cells , Triterpenes/administration & dosage , Ursolic Acid
4.
Mol Immunol ; 96: 8-18, 2018 04.
Article in English | MEDLINE | ID: mdl-29455094

ABSTRACT

Aseptic loosening due to wear particles is a serious challenge for orthopedic surgeons, sabotaging the long-term success of total joint arthroplasty. The existing treatments for aseptic loosening are still far from satisfactory, necessitating more aggressive drug exploration. Here, we examined the effect of emodin on titanium particle-induced osteolysis and further investigated its underlying mechanism in vivo and in vitro. Thirty-two C57BL/6 mice were randomly assigned into four groups: the Sham group (sham operation with vehicle injection), Vehicle group (titanium particle treatment with vehicle injections), Low group (titanium particle treatment with injections of 10 mg/kg/day emodin) and High group (titanium particle treatment with injections of 50 mg/kg/day emodin). Micro-CT scanning and histological analysis revealed that after emodin injections, the inflammatory response and bone destruction were markedly ameliorated. TRAP staining showed that osteoclast numbers were also dramatically reduced. Throughout the in vitro culture period, emodin significantly decreased the bone resorption area, number of osteoclasts and formation of F-actin rings. Mechanistic studies suggested that reduced NF-κB signaling might be mediating the inhibitory effects of emodin. Collectively, our findings suggest that emodin, a natural product extracted from Rheum palmatum, may be developed as a promising candidate for the treatment of wear particle-induced osteolysis and subsequent aseptic loosening.


Subject(s)
Emodin/pharmacology , I-kappa B Kinase/metabolism , Osteogenesis/drug effects , Osteolysis/chemically induced , Titanium/toxicity , Animals , I-kappa B Kinase/drug effects , Male , Mice , Mice, Inbred C57BL , Osteogenesis/physiology , Osteolysis/prevention & control , Phosphorylation , Prostheses and Implants , Protein Kinase Inhibitors/pharmacology , RANK Ligand/metabolism
5.
J Biol Chem ; 292(52): 21243-21252, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29097552

ABSTRACT

The spice turmeric, with its active polyphenol curcumin, has been used as anti-inflammatory remedy in traditional Asian medicine for centuries. Many cellular targets of curcumin have been identified, but how such a wide range of targets can be affected by a single compound is unclear. Here, we identified curcumin as a pro-drug that requires oxidative activation into reactive metabolites to exert anti-inflammatory activities. Synthetic curcumin analogs that undergo oxidative transformation potently inhibited the pro-inflammatory transcription factor nuclear factor κB (NF-κB), whereas stable, non-oxidizable analogs were less active, with a correlation coefficient (R2) of IC50versus log of autoxidation rate of 0.75. Inhibition of glutathione biosynthesis, which protects cells from reactive metabolites, increased the potency of curcumin and decreased the amount of curcumin-glutathione adducts in cells. Oxidative metabolites of curcumin adducted to and inhibited the inhibitor of NF-κB kinase subunit ß (IKKß), an activating kinase upstream of NF-κB. An unstable, alkynyl-tagged curcumin analog yielded abundant adducts with cellular protein that were decreased by pretreatment with curcumin or an unstable analog but not by a stable analog. Bioactivation of curcumin occurred readily in vitro, which may explain the wide range of cellular targets, but if bioactivation is insufficient in vivo, it may also help explain the inconclusive results in human studies with curcumin so far. We conclude that the paradigm of metabolic bioactivation uncovered here should be considered for the evaluation and design of clinical trials of curcumin and other polyphenols of medicinal interest.


Subject(s)
Curcumin/metabolism , Oxidation-Reduction/drug effects , Animals , Anti-Inflammatory Agents/metabolism , Curcumin/pharmacology , Glutathione/drug effects , Glutathione/metabolism , HeLa Cells , Humans , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , Mice , NF-kappa B/drug effects , NF-kappa B/metabolism , Oxidative Stress/drug effects , RAW 264.7 Cells , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
6.
Inflammation ; 40(6): 1959-1966, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28761990

ABSTRACT

In the present study, we investigated the anti-inflammatory effect of roburic acid on production of nitric oxide (NO) and interlukin-6 (IL-6) in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophage cells. We found that roburic acid reduced production of NO and IL-6, and the expression of inducible nitric oxide synthases (iNOS). Meanwhile, phosphorylation of inhibitor of κBα (IκBα) and IκB kinase α/ß (IKKα/ß), as well as translocation of nuclear factor-κB (NF-κB) to the nucleus, was suppressed by roburic acid treatment. In addition, phosphorylation of mitogen-activated protein kinase (MAPKs) including p38 and c-Jun-NH2-terminal kinase (JNK) was inhibited. Roburic acid exhibited inhibitory activities on production of NO and IL-6 via blocking IKK/IκB/NF-κB and MAPKs pathway, suggesting the potential application as a drug candidate for therapy of inflammatory diseases.


Subject(s)
Interleukin-6/biosynthesis , MAP Kinase Signaling System/drug effects , NF-kappa B/drug effects , Nitric Oxide/biosynthesis , Triterpenes/pharmacology , Animals , Gentianaceae/chemistry , I-kappa B Kinase/drug effects , I-kappa B Proteins/drug effects , Interleukin-6/antagonists & inhibitors , Lipopolysaccharides , Macrophages/metabolism , Mice , Nitric Oxide/antagonists & inhibitors , RAW 264.7 Cells
7.
Surgery ; 161(6): 1675-1682, 2017 06.
Article in English | MEDLINE | ID: mdl-28094003

ABSTRACT

BACKGROUND: Thrombomodulin, an anticoagulant that inhibits thrombin-induced growth factor promotion, also has an anti-inflammatory effect. Furthermore, thrombomodulin inhibits nuclear factor-kappa B activation, which plays a crucial role in cancer progression. We assessed the antitumor activity of recombinant thrombomodulin for pancreatic cancer. METHODS: A xenograft orthotopic model was established in mice by implantation of human pancreatic cancer cells. The animals were treated with intraperitoneal injection of recombinant thrombomodulin 5 times a week for 4 weeks. Nuclear factor-kappa B activation was evaluated by measuring nuclear localization of the p65. Efficacy of recombinant thrombomodulin on the signal transduction of nuclear factor-kappa B was measured in vitro under preconditioning with thrombin or epidermal growth factor. RESULTS: Tumor growth was suppressed by recombinant thrombomodulin (P < .05). Recombinant thrombomodulin inhibited the expression of IκB kinase ß (P < .05) and pIκBα (P < .01), as well as the activation of nuclear factor-kappa B NF-κB (P < .001). Furthermore, recombinant thrombomodulin inhibited thrombin-induced protease activate receptor 1 and nuclear factor-kappa B activation in vitro (P < .05). The number of Ki67-positive cells was decreased by recombinant thrombomodulin (P < .01). Recombinant thrombomodulin also suppressed body weight loss associated with pancreatic cancer (P < .05). No obvious adverse effects were observed. CONCLUSION: Recombinant thrombomodulin significantly suppressed tumor growth against human pancreatic cancer by blocking thrombin-induced nuclear factor-kappa B activation without adverse effects.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , I-kappa B Kinase/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Thrombomodulin/administration & dosage , Animals , Biopsy, Needle , Disease Models, Animal , Humans , I-kappa B Kinase/drug effects , Immunohistochemistry , Injections, Intraperitoneal , Male , Mice , Mice, Inbred BALB C , NF-kappa B/antagonists & inhibitors , Neoplasm Transplantation , Random Allocation , Reference Values , Signal Transduction/drug effects
8.
Osteoarthritis Cartilage ; 25(4): 589-599, 2017 04.
Article in English | MEDLINE | ID: mdl-27836674

ABSTRACT

OBJECTIVE: Osteoarthritis (OA), the most common chronic degenerative joint disease, is characterized by joint structure changes and inflammation, both mediated by the IκB kinase (IKK) signalosome complex. The ability of N-acetyl phenylalanine derivative (NAPA) to increase cartilage matrix components and to reduce inflammatory cytokines, inhibiting IKKα kinase activity, has been observed in vitro. The present study aims to further clarify the effect of NAPA in counteracting OA progression, in an in vivo mouse model after destabilization of the medial meniscus (DMM). DESIGN: 26 mice were divided into three groups: (1) DMM surgery without treatment; (2) DMM surgery treated after 2 weeks with one intra-articular injection of NAPA (2.5 mM) and (3) no DMM surgery. At the end of experimental times, both knee joints of the animals were analyzed through histology, histomorphometry, immunohistochemistry and microhardness of subchondral bone (SB) tests. RESULTS: The injection of NAPA significantly improved cartilage thickness (CT) and reduced Chambers and Mankin modified scores and fibrillation index (FI), with weaker MMP13, ADAMTS5, MMP10 and IKKα staining. The microhardness measurements did not shown statistically significant differences between the different groups. CONCLUSIONS: NAPA markedly improved the physical structure of articular cartilage while reducing catabolic enzymes, extracellular matrix (ECM) remodeling and IKKα expression, showing to be able to exert a chondroprotective activity in vivo.


Subject(s)
Cartilage, Articular/drug effects , Glucosamine/pharmacology , Knee Joint/drug effects , Osteoarthritis, Knee/immunology , Phenylalanine/analogs & derivatives , ADAMTS5 Protein/drug effects , ADAMTS5 Protein/metabolism , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Disease Models, Animal , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , Inflammation , Injections, Intra-Articular , Knee Joint/immunology , Knee Joint/metabolism , Knee Joint/pathology , Male , Matrix Metalloproteinase 10/drug effects , Matrix Metalloproteinase 10/metabolism , Matrix Metalloproteinase 13/drug effects , Matrix Metalloproteinase 13/metabolism , Menisci, Tibial/surgery , Mice , Organ Size , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Phenylalanine/pharmacology
9.
Inflammation ; 39(4): 1573-81, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27317418

ABSTRACT

Erdosteine is widely used as a mucolytic agent and also has free radical scavenging and antioxidant activities. However, little is known about the mechanisms of the anti-inflammatory effect of erdosteine. We investigated the effect of erdosteine on the activation of the nuclear factor (NF)-kB/inhibitor of NFkB (IkB), and the mitogen-activated protein kinase (MAPK) and Akt pathways in the mouse macrophage cell line RAW 264.7. Cultured RAW 264.7 cells were pretreated with erdosteine and stimulated with lipopolysaccharide (LPS). In Western blotting, pretreatment with erdosteine inhibited the IkBα degradation induced in RAW 264.7 cells by LPS. LPS-induced IkB kinase (IKK) activity and NF-kB transcription were inhibited by pretreatment with erdosteine. Production of IL-6 and IL-1ß was also inhibited by erdosteine pretreatment. However, erdosteine did not inhibit LPS-induced phosphorylation of Akt and MAPKs. These results suggest that the anti-inflammatory effect of erdosteine in mouse macrophages is mediated through inhibition of LPS-induced NF-kB activation.


Subject(s)
Thioglycolates/pharmacology , Thiophenes/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , I-kappa B Kinase/drug effects , Lipopolysaccharides/pharmacology , Mice , NF-kappa B/drug effects , NF-kappa B/metabolism , Pre-Exposure Prophylaxis , RAW 264.7 Cells
10.
Nat Commun ; 6: 6658, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25849741

ABSTRACT

The inhibitor of the nuclear factor-κB (IκB) kinase (IKK) complex is a key regulator of the canonical NF-κB signalling cascade and is crucial for fundamental cellular functions, including stress and immune responses. The majority of IKK complex functions are attributed to NF-κB activation; however, there is increasing evidence for NF-κB pathway-independent signalling. Here we combine quantitative mass spectrometry with random forest bioinformatics to dissect the TNF-α-IKKß-induced phosphoproteome in MCF-7 breast cancer cells. In total, we identify over 20,000 phosphorylation sites, of which ∼1% are regulated up on TNF-α stimulation. We identify various potential novel IKKß substrates including kinases and regulators of cellular trafficking. Moreover, we show that one of the candidates, AEG-1/MTDH/LYRIC, is directly phosphorylated by IKKß on serine 298. We provide evidence that IKKß-mediated AEG-1 phosphorylation is essential for IκBα degradation as well as NF-κB-dependent gene expression and cell proliferation, which correlate with cancer patient survival in vivo.


Subject(s)
Cell Adhesion Molecules/drug effects , Gene Expression Regulation, Neoplastic , I-kappa B Kinase/drug effects , Phosphorylation/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Blotting, Western , Cell Adhesion Molecules/metabolism , Chromatin Immunoprecipitation , Chromatography, Liquid , HEK293 Cells , Humans , I-kappa B Kinase/metabolism , I-kappa B Proteins , Immunoprecipitation , MCF-7 Cells , Mass Spectrometry , Membrane Proteins , NF-KappaB Inhibitor alpha , NF-kappa B , Phosphoproteins , RNA-Binding Proteins , Serine , Tumor Stem Cell Assay , Up-Regulation
11.
Nat Commun ; 6: 6522, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25813672

ABSTRACT

Aberrant activation of NF-κB is associated with the development of cancer and autoimmune and inflammatory diseases. IKKs are well recognized as key regulators in the NF-κB pathway and therefore represent attractive targets for intervention with small molecule inhibitors. Herein, we report that a complex natural product ainsliadimer A is a potent inhibitor of the NF-κB pathway. Ainsliadimer A selectively binds to the conserved cysteine 46 residue of IKKα/ß and suppresses their activities through an allosteric effect, leading to the inhibition of both canonical and non-canonical NF-κB pathways. Remarkably, ainsliadimer A induces cell death of various cancer cells and represses in vivo tumour growth and endotoxin-mediated inflammatory responses. Ainsliadimer A is thus a natural product targeting the cysteine 46 of IKKα/ß to block NF-κB signalling. Therefore, it has great potential for use in the development of anticancer and anti-inflammatory therapies.


Subject(s)
I-kappa B Kinase/drug effects , Lactones/pharmacology , NF-kappa B/drug effects , Neoplasms/metabolism , Sesquiterpenes/pharmacology , Signal Transduction/drug effects , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Animals , Cell Line, Tumor , Conserved Sequence , Cysteine/metabolism , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/metabolism , Lactones/metabolism , Mice , NF-kappa B/metabolism , RAW 264.7 Cells , Sesquiterpenes/metabolism
12.
Burns ; 41(5): 1076-85, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25716763

ABSTRACT

BACKGROUND: Severe burns often initiate the prevalence of hyperglycemia and insulin resistance, significantly contributing to adverse clinical outcomes. However, there are limited treatment options. This study was designed to investigate the role and the underlying mechanisms of oral antibiotics to selectively decontaminate the digestive tract (SDD) on burn-induced insulin resistance. MATERIALS AND METHODS: Rats were subjected to 40% of total body surface area full-thickness burn or sham operation with or without SDD treatment. Translocation of FITC-labeled LPS was measured at 4h after burn. Furthermore, the effect of SDD on post-burn quantity of gram-negative bacteria in gut was investigated. Serum or muscle LPS and proinflammatory cytokines were measured. Intraperitoneal glucose tolerance test and insulin tolerance test were used to determine the status of systemic insulin resistance. Furthermore, intracellular insulin signaling (IRS-1 and Akt) and proinflammatory related kinases (JNK and IKKß) were assessed by western blot. RESULTS: Burn increased the translocation of LPS from gut 4h after injury. SDD treatment effectively inhibited post-burn overgrowth of gram-negative enteric bacilli in gut. In addition, severe burns caused significant increases in the LPS and proinflammatory cytokines levels, activation of proinflammatory related kinases, and systemic insulin resistance as well. But SDD treatment could significantly attenuate burn-induced insulin resistance and improve the whole-body responsiveness to insulin, which was associated with the inhibition of gut-derived LPS, cytokines, proinflammatory related kinases JNK and IKKß, as well as activation of IRS-1 and Akt. CONCLUSIONS: SDD appeared to have an effect on proinflammatory signaling cascades and further reduced severe burn-induced insulin resistance.


Subject(s)
Anti-Infective Agents/pharmacology , Burns/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Tract/drug effects , Insulin Resistance , Animals , Blotting, Western , Burns/microbiology , Colistin/pharmacology , Cytokines/drug effects , Cytokines/metabolism , Flucytosine/pharmacology , Gastrointestinal Tract/metabolism , Glucose Tolerance Test , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , Insulin Receptor Substrate Proteins/drug effects , Insulin Receptor Substrate Proteins/metabolism , Lipopolysaccharides/metabolism , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase 4/metabolism , Male , Permeability , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Tobramycin/pharmacology
13.
Asian Pac J Cancer Prev ; 15(23): 10483-7, 2014.
Article in English | MEDLINE | ID: mdl-25556496

ABSTRACT

Heptaphylline derivatives are carbazoles in Clausena harmandiana, a medicinal plant that is utilized for headache, stomach ache, and other treatments of illness. The present study examined the effects of heptaphylline and 7-methoxyheptaphylline on apoptosis of human colon adenocarcinoma cells (HT-29 cell line). Quantification of cell viability was performed using cell proliferation assay (MTT assay) and of protein expression through immunoblotting. The results showed that only heptaphylline, but not 7-methoxyheptaphylline, significantly significantly activated cleaved of caspase-3 and poly (ADP-ribose) polymerase (PARP-1) which resulted in HT-29 cell death. We found that heptaphylline activated BH3 interacting-domain death agonist (Bid) and Bak, proapoptotic proteins. In contrast, it suppressed X-linked inhibitor-of-apoptosis protein (XIAP), Bcl-xL and survivin, inhibitors of apoptosis. In addition, heptaphylline inhibited activation of NF-κB/p65 (rel), a regulator of apoptotic regulating proteins by suppressing the activation of Akt and IKKα, upstream regulators of p65. The findings suggested that heptaphylline induces apoptosis in human colon adenocarcinoma cells .


Subject(s)
Adenocarcinoma/metabolism , Apoptosis/drug effects , Carbazoles/pharmacology , Colonic Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Transcription Factor RelA/drug effects , BH3 Interacting Domain Death Agonist Protein/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , HT29 Cells , Humans , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , Inhibitor of Apoptosis Proteins/drug effects , Inhibitor of Apoptosis Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Survivin , Transcription Factor RelA/metabolism , X-Linked Inhibitor of Apoptosis Protein/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-2 Homologous Antagonist-Killer Protein/drug effects , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/drug effects , bcl-X Protein/metabolism
14.
Oral Oncol ; 49(12): 1113-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24075781

ABSTRACT

OBJECTIVES: Ikappa B kinase alpha (IKKα) plays an inhibitory role in the development of epithelial-derived tumors. However, its specific function in the development of nasopharyngeal carcinoma (NPC) remains unknown. In this study we identify the role and mechanism of IKKα in IKKα-mediated NPC development. MATERIAL AND METHODS: The effect of IKKα on migration, invasion and tumorigenesis of NPC cell lines was determined using in vitro and in vivo studies. SUNE-1-5-8F cells transfected to overexpress IKKα, SUNE-1-6-10B cells with shRNA-mediated knockdown of IKKα, and three NPC cell lines were studied using Western blotting techniques to compare the major molecules in NF-κB pathways. Additionally, the extracellular signal-regulated kinase (ERK) pathway and matrix metalloproteinases (MMPs) in IKKα-regulated NPC and the effect of Epstein-Barr Nuclear Antigen 1 (EBNA1) on IKKα were examined. RESULTS: IKKα was underexpressed in highly invasive SUNE-1-5-8F cells compared with non-invasive cells (SUNE-1 and SUNE-6-10B). Overexpression of IKKα in SUNE-1-5-8F cells was achieved through transfection and resulted in inhibited migration and invasion in vitro. Furthermore, IKKα inhibited tumorigenesis in mice inoculated with IKKα-transfected NPC cells in vivo. These processes were independent of the conventional effect of IKKα on Nuclear factor κB (NF-κB) pathways. The ERK pathway was involved in IKKα-related NPC inhibition. Phosphorylation of ERK1/2 and subsequent secretion of MMP-9 were inhibited by the ERK inhibitor U0126 and not regulated by overexpressed IKKα. EBNA1 knockdown using small interfering RNA (siRNA) did not alter the expression of IKKα. CONCLUSION: Increase in IKKα expression suppresses the progression of NPC through a NF-κB-independent and ERK-dependent pathway.


Subject(s)
I-kappa B Kinase/metabolism , Nasopharyngeal Neoplasms/metabolism , Animals , Blotting, Western , Carcinoma , Cell Line, Tumor , Epstein-Barr Virus Nuclear Antigens/pharmacology , Humans , I-kappa B Kinase/drug effects , MAP Kinase Signaling System/physiology , Male , Matrix Metalloproteinases/metabolism , Mice , NF-kappa B/metabolism , Nasopharyngeal Carcinoma , Phosphorylation
15.
Br J Haematol ; 161(1): 57-67, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23356405

ABSTRACT

Heat shock protein 90 (HSP90; HSP90AA1) is a molecular chaperone involved in signalling pathways for cell proliferation, survival, and cellular adaptation. Inhibitors of HSP90 are being examined as anti-cancer agents, but the critical molecular mechanism(s) of their activity remains unresolved. HSP90 inhibition potentially facilitates the simultaneous targeting of multiple molecules within tumour cells and represents an attractive therapeutic proposition. Here, we investigated HSP90 as a molecular target for acute myeloid leukaemia (AML) using the novel HSP90 inhibitor NVP-AUY922-AG. NVP-AUY922-AG induced dose-dependent killing in myeloid cell lines and primary AML blasts. In primary blasts, cell death in response to NVP-AUY922-AG was seen at concentrations almost 2 logs lower than cytarabine (Ara-C) (50% lethal dose = 0·12 µ mol/l ± 0·28). NVP-AUY922-AG was significantly less toxic to normal bone marrow (P = 0·02). In vitro response to NVP-AUY922-AG did not correlate with response to Ara-C (r(2) = 0·0006). NVP-AUY922-AG was highly synergistic with Ara-C in cell lines and in 20/25 of the primary samples tested. NVP-AUY922-AG induced increases in HSP70 expression and depletion of total AKT, IKKα and IKKß in cell lines and primary blasts. This study shows that the novel HSP90 inhibitor NVP-AUY922-AG has significant single agent activity in AML cells and is synergistic with Ara-C.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Resorcinols/pharmacology , Adult , Aged , Aged, 80 and over , Annexin A5/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Cytarabine/administration & dosage , Cytarabine/pharmacology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Drug Synergism , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , Isoxazoles/administration & dosage , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Resorcinols/administration & dosage , Signal Transduction/drug effects , Tumor Cells, Cultured/drug effects , Young Adult
16.
Am J Physiol Heart Circ Physiol ; 303(12): H1435-45, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23042949

ABSTRACT

Pressure overload is known to be a cause of cardiac hypertrophy that often transits to heart failure. Although nuclear factor (NF)-κB is a key factor in the progression of cardiac hypertrophy, its pathophysiology is yet to be elucidated. Thus, we aimed to show that inhibition of NF-κB activation improves pressure overload-induced cardiac dysfunction. To assess the effect of inhibition on NF-κB activation in pressure overload cardiac hypertrophy, we used IMD-1041 in a murine thoracic aortic constriction (TAC) model. IMD-1041 inhibits the phosphorylation of IκB via inhibition of IκB kinase-ß. IMD-1041 (100 mg·kg(-1)·day(-1)) or vehicle was administered orally into mice once a day, and mice were euthanized on day 42 after TAC. TAC resulted in left ventricular wall thickening, cardiac dysfunction, and increases of heart and lung weight, whereas IMD-1041 significantly suppressed the development of cardiac hypertropy 6 wk after TAC. Histologically, developed cardiac fibrosis and cardiomyocyte hypertrophy occurred in the vehicle-treated group, whereas IMD-1041 significantly attenuated these changes. IMD-1041 suppressed the expression of p65-positive cells and nuclear translocation of p65 induced by TAC compared with vehicle. Matrix metalloproteinase-2 activity increased in the vehicle + TAC-treated group; however, it was suppressed in the IMD-1041 + TAC-treated group. IMD-1041 treatment from day 28 to day 42 after TAC significantly attenuated the decrease in the percentage of fractional shortening and cardiac fibrosis without an antihypertrophic effect. In conclusion, IMD-1041 may be useful for preventing pressure overload-induced cardiac dysfunction and the transition of cardiac hypertrophy to contraction failure via suppression of NF-κB activation.


Subject(s)
Aorta, Thoracic/physiopathology , Enzyme Inhibitors/pharmacology , Heart/drug effects , Heart/physiopathology , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Vasoconstriction/physiology , Animals , Apoptosis/drug effects , Benzamides/pharmacology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Disease Models, Animal , Fibrosis/prevention & control , Heart Ventricles/drug effects , Heart Ventricles/pathology , I-kappa B Kinase/drug effects , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , NF-kappa B/metabolism , Phosphorylation
17.
J Periodontal Res ; 47(2): 204-11, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21972936

ABSTRACT

BACKGROUND AND OBJECTIVE: Host modulatory agents directed at inhibiting specific proinflammatory mediators could be beneficial in terms of attenuating periodontal disease progression and potentially enhancing therapeutic responses. The aim of this study was to investigate whether daidzein could modulate the production inflammatory mediators in macrophages stimulated with lipopolysaccharide (LPS) from Prevotella intermedia, a pathogen implicated in periodontal disease, and to delineate underlying mechanisms of action. MATERIAL AND METHODS: LPS was extracted from P. intermedia ATCC 25611 cells by the standard hot phenol-water method. The amounts of nitric oxide (NO) and interleukin-6 (IL-6) secreted into the culture medium were assayed. A real-time PCR was performed to quantify inducible nitric oxide synthase (iNOS) and IL-6 mRNA expression. We used immunoblot analysis to characterize iNOS protein expression, phosphrylation of c-Jun N-terminal kinase (JNK) and p38, degradation of inhibitory κB-α (IκB-α), nuclear translocation of nuclear factor-κB (NF-κB) subunits and phosphorylation of signal transducer and activator of transcription 1 (STAT1). The DNA-binding activity of NF-κB was assessed by using ELISA-based kits. RESULTS: Daidzein significantly inhibited the production of NO and IL-6, as well as their mRNA expression, in P. intermedia LPS-treated RAW264.7 cells. The JNK and p38 pathways were not involved in the regulation of LPS-induced NO and IL-6 release by daidzein. Daidzein inhibited the degradation of IκB-α induced by P. intermedia LPS. In addition, daidzein suppressed NF-κB transcriptional activity via regulation of the nuclear translocation and DNA-binding activity of NF-κB p50 subunit and blocked STAT1 phosphorylation. CONCLUSION: Although additional studies are required to dissect the molecular mechanism of action, our results suggest that daidzein could be a promising agent for treating inflammatory periodontal disease. Further research in animal models of periodontitis is necessary to better evaluate the potential of daidzein as a novel therapeutic agent to treat periodontal disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Growth Inhibitors/pharmacology , Isoflavones/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Phytoestrogens/pharmacology , Prevotella intermedia , Animals , Bacteriological Techniques , Cell Culture Techniques , Cell Line , I-kappa B Kinase/drug effects , Inflammation Mediators/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/drug effects , Janus Kinase 2/drug effects , Mice , NF-kappa B/drug effects , NF-kappa B p50 Subunit/drug effects , Nitric Oxide Synthase Type II/drug effects , Phosphorylation , STAT1 Transcription Factor/drug effects , Transcription Factor RelA/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects
18.
Int J Med Mushrooms ; 13(1): 19-25, 2011.
Article in English | MEDLINE | ID: mdl-22135900

ABSTRACT

Breast cancer is the most commonly diagnosed cancer among women. Currently, there is no effective therapy for malignant estrogen-independent breast cancer. In our study, we used hydrogen peroxide, a well-known strong oxidative reagent capable of activating the nuclear factor kappa B (NF-kappaB) transcription factor. The IC50 value of the culinary-medicinal Shaggy Inc Cap mushroom Coprinus comatus culture liquid crude extract on MCF7 cell viability was found to be as low as 76 microg/mL, and the IC50 value of C. comatus ethyl acetate extract was only 32 microg/ mL. Our results also showed that both extracts significantly affected IkappaBalpha phosphorylation in a dose-dependent manner. The effect of ethyl acetate extract was comparable to the effect of curcumin, a known NF-kappaB pathway inhibitor, and seemed to be the most active inhibitor of H2O2-dependent IkappaBalpha phosphorylation. In addition, the data obtained showed that only ethyl acetate extract inhibited the activity of IKK complex, at close to 90% as compared to the control of the untreated sample. These results suggest that C. comatus contains potent compounds capable of inhibiting NF-kappaB function and also possibly acts as an antitumor agent.


Subject(s)
Complex Mixtures/pharmacology , Coprinus/chemistry , Hydrogen Peroxide/pharmacology , I-kappa B Kinase/drug effects , I-kappa B Proteins/drug effects , NF-kappa B/drug effects , Acetates/chemistry , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival , Complex Mixtures/isolation & purification , Dose-Response Relationship, Drug , Female , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , I-kappa B Proteins/metabolism , Inhibitory Concentration 50 , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Time Factors
19.
Oral Oncol ; 47(9): 818-26, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21745758

ABSTRACT

The transcriptional activation of NF-κB signalling has been identified as a major pathway involved in inflammation and tumor aggressiveness in a number of human cancers. Here we identify the impact of miscellaneous known and so far unknown NF-κB inhibitors originating from different drug classes on the function and proliferation of HNSCC. In detail: HNSCC cell lines were exposed to Acetylsalicylic Acid (ASA), Celecoxib, Dexamethasone, Curcumin and EPs 7630. Our major interest was to detect upstream alterations in cell signalling after applying NF-κB inhibiting substances. The inhibition of NF-κB signalling leads to an upstream regulation of Toll-like-receptor 3 (TLR3), a predominant receptor driving cell expansion. We find a marked downregulation of TLR3 and IKK complex, documenting upstream responses to NF-κB inhibition by every agent tested. In a second step we further analyse proliferation, cytokine production and alterations in the expression of downstream proteins such as cyclin D1 and c-Myc. Our data demonstrate decreased proliferation in response to incubation with aforementioned agents. Modulation of TLR3 and NF-κB expression is accompanied by altered profiles of IL-6 and IL-8 which are relevant cytokines in HNSCC progression. Proto-oncogenes cyclin D1 and c-myc are downregulated by all substances. Apart from the interplay of cytokines and TLR3, we substantiated EPs 7630 as a new and natural NF-κB inhibitor.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/metabolism , Cyclooxygenase Inhibitors/pharmacology , Hypopharyngeal Neoplasms/metabolism , NF-kappa B/antagonists & inhibitors , Toll-Like Receptor 3/metabolism , Cell Proliferation/drug effects , Cyclin D/metabolism , Cytokines/metabolism , Humans , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Male , Middle Aged , NF-kappa B/drug effects , NF-kappa B/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Toll-Like Receptor 3/drug effects
20.
Cancer Biol Ther ; 12(1): 59-68, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21532339

ABSTRACT

Gallotannin (GT), the polyphenolic hydrolyzable tannin, exhibits anti-inflammatory and anticancer activities through mechanisms that are not fully understood. Several effects modulated by GT have been shown to be linked to interference with inflammatory mediators. Considering the central role of nuclear factor kappa B (NF-ĸB) in inflammation and cancer, we investigated the effect of GT on NF-ĸB signaling in HT-29 and HCT-116 human colon cancer cells. DNA binding assays revealed significant suppression of tumor necrosis factor (TNF-α)-induced NFĸB activation which correlated with the inhibition of IĸBα phosphorylation and degradation. Sequentially, p65 nuclear translocation and DNA binding were inhibited. GT also down-regulated the expression of NFĸB-regulated inflammatory cytokines (IL-8, TNF-α, IL-1α) and caused cell cycle arrest and accumulation of cells in pre-G 1 phase. In vivo, GT (25 mg/kg body weight) injected intraperitoneally (i.p.) prior to or after tumor inoculation significantly decreased the volume of human colon cancer xenografts in NOD/SCID mice. GT-treated xenografts showed significantly lower microvessel density (CD31) as well as lower mRNA expression levels of IL-6, TNF-α and IL-1α and of the proliferation (Ki-67) and angiogenesis (VEGFA) proteins, which may explain GTs in vivo anti-tumorigenic effects. Overall, our results indicate that the anti-inflammatory and antitumor activities of GT may be mediated in part through the suppression of NF-ĸB activation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colonic Neoplasms/drug therapy , Hydrolyzable Tannins/pharmacology , NF-kappa B p50 Subunit/antagonists & inhibitors , NF-kappa B p50 Subunit/metabolism , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Cell Cycle/drug effects , Cell Line, Tumor , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytokines/metabolism , Female , G1 Phase/drug effects , Humans , Hydrolyzable Tannins/administration & dosage , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Interleukin-1alpha/genetics , Interleukin-6/genetics , Mice , Mice, Inbred NOD , Microvessels/drug effects , Microvessels/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , RNA, Messenger , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL