Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Cancer Biol Ther ; 25(1): 2322206, 2024 12 31.
Article in English | MEDLINE | ID: mdl-38436092

ABSTRACT

Salidroside inhibited the proliferation of cancer cell. Nevertheless, the mechanism has not been completely clarified. The purpose of the study is to explore the mechanisms of salidroside against gastric cancer. To analyze the changes of microRNA (miRNA) in gastric cancer cells under the treatment of salidroside, the miRNA expression was analyzed by using RNA-seq in cancer cells for 24 h after salidroside treatment. The differentially expressed miRNAs were clustered and their target genes were analyzed. Selected miRNA and target mRNA genes were further verified by q-PCR. The expressions of target genes in cancer cells were detected by immunohistochemistry. Cancer cell apoptotic index was significantly increased after salidroside treatment. The proliferation of gastric cancer cells were blocked at S-phase cell cycle. The expression of 44 miRNAs changed differentially after salidroside treatment in cancer cells. Bioinformatic analysis showed that there were 1384 target mRNAs corresponding to the differentially expressed miRNAs. Surprisingly, salidroside significantly up-regulated the expression of tumor suppressor miR-1343-3p, and down-regulated the expression of MAP3K6, STAT3 and MMP24-related genes. Salidroside suppressed the growth of gastric cancer by inducing the cancer cell apoptosis, arresting the cancer cell cycle and down-regulating the related signal transduction pathways. miRNAs are expressed differentially in gastric cancer cells after salidroside treatment, playing important roles in regulating proliferation and metastasis. Salidroside may suppress the growth of gastric cancer by up-regulating the expression of the tumor suppressor miR-1343-3p and down-regulating the expression of MAP3K6 and MMP24 signal molecules.


Subject(s)
Glucosides , MicroRNAs , Phenols , Stomach Neoplasms , Humans , Cell Proliferation , Matrix Metalloproteinases, Membrane-Associated , MicroRNAs/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Kinase Kinases/metabolism
2.
Molecules ; 26(10)2021 May 20.
Article in English | MEDLINE | ID: mdl-34065429

ABSTRACT

Barringtonia augusta methanol extract (Ba-ME) is a folk medicine found in the wetlands of Thailand that acts through an anti-inflammatory mechanism that is not understood fully. Here, we examine how the methanol extract of Barringtonia augusta (B. augusta) can suppress the activator protein 1 (AP-1) signaling pathway and study the activities of Ba-ME in the lipopolysaccharide (LPS)-treated RAW264.7 macrophage cell line and an LPS-induced peritonitis mouse model. Non-toxic concentrations of Ba-ME downregulated the mRNA expression of cytokines, such as cyclooxygenase and chemokine ligand 12, in LPS-stimulated RAW264.7 cells. Transfection experiments with the AP-1-Luc construct, HEK293T cells, and luciferase assays were used to assess whether Ba-ME suppressed the AP-1 functional activation. A Western blot assay confirmed that C-Jun N-terminal kinase is a direct pharmacological target of Ba-ME action. The anti-inflammatory effect of Ba-ME, which functions by ß-activated kinase 1 (TAK1) inhibition, was confirmed by using an overexpression strategy and a cellular thermal shift assay. In vivo experiments in a mouse model of LPS-induced peritonitis showed the anti-inflammatory effect of Ba-ME on LPS-stimulated macrophages and acute inflammatory mouse models. We conclude that Ba-ME is a promising anti-inflammatory drug targeting TAK1 in the AP-1 pathway.


Subject(s)
Barringtonia/chemistry , MAP Kinase Kinase Kinases/drug effects , Plant Extracts/pharmacology , Transcription Factor AP-1/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Blotting, Western , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/toxicity , Macrophages/drug effects , Methanol/chemistry , Mice , Peritonitis/chemically induced , Peritonitis/prevention & control , RAW 264.7 Cells
3.
Aging (Albany NY) ; 13(4): 5150-5163, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33535169

ABSTRACT

The Nicotinamide phosphoribosyltransferase (Nampt)-NAD-Sirt1 pathway modulates processes involved in the pathogenesis of multiple diseases by influencing inflammation. This study aimed to explore the effect of Nampt in osteogenic differentiation and inflammatory response of osteoblastic MC3T3-E1 cells. We developed an in vitro model of lipopolysaccharide (LPS)-induced inflammation and showed that Nampt and Sirt1 were significantly upregulated in LPS-treated MC3T3-E1 cells. LPS induced secretion of the proinflammatory cytokine interleukin-6 (IL-6) and attenuated osteogenic differentiation. Then we transfected cells with adenoviruses to knock down or over express Nampt. Nampt promoted the expression of IL-6, TAK1 and phospho-NF-κB p65 after LPS treatment. Overexpression of Nampt overrode the effect of LPS and rescued LPS-induced inhibition on osteogenic differentiation. FK866, a Nampt inhibitor, had the same inhibitory effect as Nampt knockdown. In addition, Sirt1 suppression by EX527 decreased IL-6 secretion and NF-κB activation without changing the level of Nampt. EX527 also decreased osteogenic differentiation. Incubation with NMN or SRT 1720 also counteract the inhibitory effect of LPS and rescued osteoblast differentiation. Therefore, we demonstrated that Nampt acted both in promoting osteoblast differentiation and in enhancing inflammatory response, mediated by Sirt1 in MC3T3-E1 cells.


Subject(s)
Cell Differentiation/genetics , Cytokines/genetics , Interleukin-6/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Osteoblasts/metabolism , Osteogenesis/genetics , Sirtuin 1/genetics , Acrylamides/pharmacology , Animals , Carbazoles/pharmacology , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Gene Knockdown Techniques , Inflammation , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Kinase Kinases/metabolism , Mice , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Piperidines/pharmacology , Signal Transduction , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/metabolism , Transcription Factor RelA/drug effects , Transcription Factor RelA/metabolism
4.
Int J Neurosci ; 131(10): 975-983, 2021 Oct.
Article in English | MEDLINE | ID: mdl-32378973

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is the most common aggressive primary cancer occurring in the brain tissue. GBM accounts 16% of primary brain tumors and half of gliomas. Additionally, the incidence of GBM is increases with aging, and reaches the peak at the age of 75 to 84 years. The survival of patients with GBM remains at a low level, only less than 5% patients diagnosed with GBM survive for 5 years. Temozolomide (TMZ) is a DNA alkylating agent and is currently a first line chemotherapeutic treatment for GBM. TMZ combined with radiation therapy has been shown to prolong the overall survival (OS) to 14.6 months compared with 12.1 months for radiation therapy alone. NF-E2-related factor 2 (Nrf2) is a transcription factor that contains seven functional domains. The binding of Keap1 to Nrf2 is a central regulator of the cellular defense mechanism against environmental stresses. METHODS: First, Nrf2 overexpression and inhibition models were constructed in U251 cells using transfection. The percentage of viable cells was detected using the MTT assay. Then, the expression of the HO-1 regulator was detected using qPCR, and the concentrations of oxidative stress related factors were detected using ELISAs. The levels of proteins related to oxidative stress and the Ras/Raf/MEK signaling pathway was detected using western blotting analysis. RESULTS: We initially established Nrf2 inhibition and activation cell models in U251 cells and found that the inhibition of Nrf2 expression decreased the mRNA and protein levels of the anti-oxidative enzymes, as well as the secretion of these enzymes into the cellular microenvironment. These effects might be mediated by the inhibition of Ras/Raf/MEK signaling pathway, leading to the inhibition of cellular proliferation. CONCLUSIONS: Inhibition of Nrf2 expression might enhance the effect of TMZ on the treatment of GBM and might be a new therapeutic strategy.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Glioma/drug therapy , MAP Kinase Kinase Kinases/drug effects , NF-E2-Related Factor 2/drug effects , Temozolomide/pharmacology , raf Kinases/drug effects , ras Proteins/drug effects , Humans , MAP Kinase Kinase Kinases/antagonists & inhibitors , NF-E2-Related Factor 2/antagonists & inhibitors , Signal Transduction/drug effects , raf Kinases/antagonists & inhibitors , ras Proteins/antagonists & inhibitors
5.
Int J Mol Sci ; 21(9)2020 May 09.
Article in English | MEDLINE | ID: mdl-32397561

ABSTRACT

Although various advancements in radical surgery and neoadjuvant chemotherapy have been developed in treating osteosarcoma (OS), their clinical prognosis remains poor. A synthetic chemical compound, 3-hydroxylflavone, that is reported to regulate ROS production is known to inhibit human bone osteosarcoma cells. However, its role and mechanism in human OS cells remains unclear. In this study, we have determined the potential of 3-Hydroxy-2-phenylchromone (3-HF) against OS using human osteosarcoma (HOS) cells. Our previous studies showed that Zipper sterile-alpha-motif kinase (ZAK), a kinase member of the MAP3K family, was involved in various cellular events such as cell proliferation and cell apoptosis, and encoded two transcriptional variants, ZAKα and ß. In this study, we show that 3-HF induces the expression of ZAK and thereby enhances cellular apoptosis. Using gain of function and loss of function studies, we have demonstrated that ZAK activation by 3-HF in OS cells is confined to a ZAKß form that presumably plays a leading role in triggering ZAKα expression, resulting in an aggravated cancer apoptosis. Our results also validate ZAKß as the predominant form of ZAK to drive the anticancer mechanism in HOS cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/pathology , Flavonoids/pharmacology , MAP Kinase Kinase 4/drug effects , MAP Kinase Kinase Kinases/drug effects , Osteosarcoma/pathology , Caspase 3/biosynthesis , Caspase 3/genetics , Cell Line, Tumor , Enzyme Activation/drug effects , Gain of Function Mutation , Humans , Loss of Function Mutation , MAP Kinase Kinase Kinases/genetics , Membrane Potential, Mitochondrial/drug effects , Protein Isoforms/drug effects , Protein Isoforms/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
6.
Int J Mol Sci ; 21(6)2020 Mar 16.
Article in English | MEDLINE | ID: mdl-32187984

ABSTRACT

Various herbal extracts containing luteolin-7-O-glucuronide (L7Gn) have been traditionally used to treat inflammatory diseases. However, systemic studies aimed at elucidating the anti-inflammatory and anti-oxidative mechanisms of L7Gn in macrophages are insufficient. Herein, the anti-inflammatory and anti-oxidative effects of L7Gn and their underlying mechanisms of action in macrophages were explored. L7Gn inhibited nitric oxide (NO) production in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages by transcriptional regulation of inducible NO synthase (iNOS) in a dose-dependent manner. The mRNA expression of inflammatory mediators, including cyclooxygenase-2 (COX-2), interleukin-6 (IL-6), IL-1ß, and tumor necrosis factor-α (TNF-α), was inhibited by L7Gn treatment. This suppression was mediated through transforming growth factor beta-activated kinase 1 (TAK1) inhibition that leads to reduced activation of nuclear factor-κB (NF-κB), p38, and c-Jun N-terminal kinase (JNK). L7Gn also enhanced the radical scavenging effect and increased the expression of anti-oxidative regulators, including heme oxygenase-1 (HO-1), glutamate-cysteine ligase catalytic subunit (GCLC), and NAD(P)H quinone oxidoreductase 1 (NQO1), by nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) activation. These results indicate that L7Gn exhibits anti-inflammatory and anti-oxidative properties in LPS-stimulated murine macrophages, suggesting that L7Gn may be a suitable candidate to treat severe inflammation and oxidative stress.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Lipopolysaccharides/adverse effects , Luteolin/antagonists & inhibitors , MAP Kinase Kinase Kinases/drug effects , Macrophages/drug effects , NF-E2-Related Factor 2/drug effects , Animals , Cell Survival/drug effects , Cyclooxygenase 2/metabolism , Heme Oxygenase-1/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Luteolin/chemistry , Luteolin/pharmacology , MAP Kinase Kinase Kinases/metabolism , Macrophages/metabolism , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/metabolism
7.
Neurochem Int ; 122: 59-72, 2019 01.
Article in English | MEDLINE | ID: mdl-30423425

ABSTRACT

Midazolam and ketamine-induced anesthesia were recently shown to induce a disruption of MEK/ERK sequential phosphorylation with parallel upregulation of p-FADD in the mouse brain. The present study was designed to assess whether other structurally diverse anesthetic agents (pentobarbital, ethanol, chloral hydrate, isoflurane) also impair brain p-MEK to p-ERK signal and increase p-FADD during the particular time course of 'sleep' in mice. Pentobarbital (50 mg/kg)-, ethanol (4000 mg/kg)-, chloral hydrate (400 mg/kg)-, and isoflurane (2% in O2)-induced anesthesia (range: 24-60 min) were associated with unaltered or increased p-MEK1/2 (up to +155%) and decreased p-ERK1/2 (up to -60%) contents, revealing disruption of MEK to ERK activation in mouse brain cortex. These anesthetic agents also upregulated cortical p-FADD (up to +110%), but not total FADD (moderately decreased), which resulted in increased neuroplastic/survival p-FADD/FADD ratios (up to +2.8 fold). The inhibition of pentobarbital metabolism with SKF525-A (a cytochrome P450 inhibitor) augmented barbiturate anesthesia (2.6 times) and induced a greater and sustained upregulation of p-MEK with p-ERK downregulation, as well as prolonged increases of p-FADD content and p-FADD/FADD ratio (effects lasting for more than 240 min). Pentobarbital also upregulated significantly the cortical contents of other markers of neuroplasticity such as the ERK inhibitor p-PEA-15 (up to +46%), the transcription factor NF-κB (up to +27%) and the synaptic density protein PSD-95 (up to +20%) during 'sleep'. The results reveal a paradoxical stimulation of p-MEK without the concomitant (canonical) activation of p-ERK (e.g. with pentobarbital and isoflurane), for which various molecular mechanisms are discussed. The downregulation of brain p-ERK may participate in the manifestations of adverse effects displayed by most hypnotic/anesthetic agents in clinical use (e.g. amnesia).


Subject(s)
Brain/drug effects , Fas-Associated Death Domain Protein/metabolism , MAP Kinase Kinase Kinases/drug effects , Pentobarbital/pharmacology , Anesthetics/pharmacology , Animals , Brain/metabolism , Hypnotics and Sedatives/pharmacology , Ketamine/pharmacology , Male , Mice , Neuronal Plasticity/drug effects , Transcriptional Activation/drug effects , Up-Regulation/drug effects
9.
Rheumatology (Oxford) ; 56(7): 1060-1068, 2017 07 01.
Article in English | MEDLINE | ID: mdl-27550296

ABSTRACT

Pro-inflammatory cytokines such as IL-1ß, IL-6 and TNF-α are central regulators of autoinflammatory diseases. While targeting these cytokines has proven to be a successful clinical strategy, the long-term challenges such as drug resistance, lack of efficacy and poor clinical outcomes in some patients are some of the limitations faced by these therapies. This has ignited strategies to reduce inflammation by potentially targeting a variety of molecules, including cell surface receptors, signalling proteins and/or transcription factors to minimize cytokine-induced inflammation and tissue injury. In this regard, transforming growth factor ß activated kinase 1 (TAK1) is activated in the inflammatory signal transduction pathways in response to IL-1ß, TNF-α or toll-like receptor stimulation. Because of its ideal position upstream of mitogen-activated protein kinases and the IκB kinase complex in signalling cascades, targeting TAK1 may be an attractive strategy for treating diseases characterized by chronic inflammation. Here, we discuss the emerging role of TAK1 in mediating the IL-1ß, TNF-α and toll-like receptor mediated inflammatory responses in diseases such as RA, OA, gout and SS. We also review evidence suggesting that TAK1 inhibition may have potential therapeutic value. Finally, we focus on the current status of the development of TAK1 inhibitors and suggest further opportunities for testing TAK1 inhibitors in rheumatic diseases.


Subject(s)
Cytokines/drug effects , MAP Kinase Kinase Kinases/drug effects , Molecular Targeted Therapy , Rheumatic Diseases/drug therapy , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism , Antirheumatic Agents/therapeutic use , Cytokines/metabolism , Female , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , MAP Kinase Kinase Kinases/metabolism , Male , Prognosis , Rheumatic Diseases/blood , Rheumatic Diseases/diagnosis
10.
Hepatology ; 65(4): 1222-1236, 2017 04.
Article in English | MEDLINE | ID: mdl-27863449

ABSTRACT

Hepatocellular carcinoma (HCC) is the second-leading cause of cancer-related deaths worldwide with a poor survival rate. As many as 40% of HCCs are clonal, with alteration of key tumor-suppressor pathways in stem cells as the primary cause of HCC initiation. However, mechanisms that generate metastatic stem cells in preneoplastic liver tissue are not well understood. We hypothesized that chronic inflammation is a major driver of the transformation of genetically defective liver stem cells (LSCs) into highly metastatic liver cancer cells in premalignant liver tissue. We developed models of chronic inflammation in wild-type (WT) and ß2-spectrin (ß2SP)+/- (SPTBN1) mice. CD133+ LSCs derived from preneoplastic livers of ß2SP+/- mice treated with interleukin-6 (pIL6; IL6 ß2SP+/- LSCs) were highly tumorigenic and metastatic, whereas those derived from WT mice treated with pIL6 (IL6 WT LSCs) had significantly less proliferation and no tumorigenic properties. IL6 ß2SP+/- LSCs not only exhibited nuclear localization of Twist and Slug, markers of epithelial-mesenchymal transition (EMT), but also constitutive activation of nuclear factor kappa B (NFκB; RELA). Knockdown of NFκB decreased the EMT phenotypes and metastatic capacity of these cells. NFκB in IL6 ß2SP+/- LSCs was activated by transforming growth factor ß (TGFß)-activated kinase 1 (TAK1; MAP3K7), which is associated with poor survival in HCC and interleukin-6 (IL6) expression. The amount of constitutively activated NFκB increased dramatically from normal to cirrhotic to HCC tissues from human patients. CONCLUSION: IL6-mediated inflammation programs constitutive activation of the TAK1-NFκB signaling cascade in CD133+ LSCs, and this program interacts with deficient TGFß signaling, thereby accelerating the transformation of normal LSCs to metastatic cancer stem cells (mCSCs). Indeed, this study delineates the development of EMT-positive mCSCs in HCC-free liver tissue upon chronic inflammation. (Hepatology 2017;65:1222-1236).


Subject(s)
Carcinoma, Hepatocellular/pathology , Interleukin-6/pharmacology , Liver Neoplasms/pathology , MAP Kinase Kinase Kinases/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Biopsy, Needle , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Immunohistochemistry , Liver Neoplasms/metabolism , MAP Kinase Kinase Kinases/drug effects , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Random Allocation , Statistics, Nonparametric , Transforming Growth Factor beta1/drug effects
11.
Toxicol Lett ; 264: 87-98, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27836799

ABSTRACT

Cucurbitacin-I, a natural triterpenoids initially identified in medicinal plants, shows a potent anticancer effect on a variety of cancer cell types. Nevertheless, the cardiotoxicity of cucurbitacin-I has not heretofore been reported. In this study, the mechanisms of cucurbitacin-I-induced cardiotoxicity were examined by investigating the role of MAPK-autophagy-dependent pathways. After being treated with 0.1-0.3µM cucurbitacin-I for 48h, H9c2 cells showed a gradual decrease in the cell viabilities, a gradual increase in cell size, and mRNA expression of ANP and BNP (cardiac hypertrophic markers). Cucurbitacin-I concentration-dependent apoptosis of H9c2 cells was also observed. The increased apoptosis of H9c2 cells was paralleling with the gradually strong autophagy levels. Furthermore, an autophagy inhibitor, 3-MA, was used to block the cucurbitacin-I-stirred autophagy, and then the hypertrophy and apoptosis induced by 0.3µM cucurbitacin-I were significantly attenuated. In addition, cucurbitacin-I exposure also activated the MAPK signaling pathways, including ERK1/2, JNK, and p38 kinases. Interestingly, only the ERK inhibitor U0126, but not the JNK inhibitor SP600125 and p38 MAPK inhibitor SB203580, weakened the induction of 0.3µM cucurbitacin-I in hypertrophy, autophagy and apoptosis. Our findings suggest that cucurbitacin-I can increase the autophagy levels of H9c2 cells, most likely, through the activation of an ERK-autophagy dependent pathway, which results in the hypertrophy and apoptosis of cardiomyocytes.


Subject(s)
Autophagy/drug effects , Cardiomegaly/chemically induced , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Signaling System/drug effects , Myoblasts, Cardiac/drug effects , Triterpenes/pharmacology , Atrial Natriuretic Factor/biosynthesis , Cardiomegaly/pathology , Cell Size/drug effects , Cell Survival/drug effects , Humans , Imidazoles/pharmacology , MAP Kinase Kinase 4/antagonists & inhibitors , Natriuretic Peptide, Brain/biosynthesis , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects
12.
Expert Opin Ther Pat ; 26(5): 607-16, 2016 May.
Article in English | MEDLINE | ID: mdl-27043251

ABSTRACT

INTRODUCTION: The dual leucine zipper kinase (DLK, MAP3K12) is essential for neuronal development and has been shown to mediate axon regeneration. On the other hand, DLK is involved in the pathogenesis of neurodegenerative disease and diabetes mellitus. Several patents have been published claiming to modulate or inhibit DLK by various approaches including ATP competitive inhibitors. In addition, two publications describe SAR of highly selective DLK inhibitors with efficacy in distinct mouse models of neurodegeneration. AREAS COVERED: This review summarized patents claiming to modulate DLK activity published between 2010 and 2015. Peer-reviewed publications related to the patents and additional peer-reviewed publications are included. This article describes 18 patents from three pharmaceutical companies and three academic research groups. EXPERT OPINION: Several methods are proposed to modulate DLK activity, some of them very experimental and not suitable for easy application in patients. ATP competitive kinase inhibitors exert high affinity, but for the majority, no information about their selectivity is available. To date, two inhibitors have been tested in mice. Given the controversial findings that DLK is required for neurodegeneration and for axon regeneration, more research is needed to further elucidate the regulation and the function of this kinase in diverse organs/tissues and under physiological and pathological conditions.


Subject(s)
Drug Design , MAP Kinase Kinase Kinases/drug effects , Neurodegenerative Diseases/drug therapy , Animals , Axons/metabolism , Disease Models, Animal , Humans , MAP Kinase Kinase Kinases/metabolism , Mice , Nerve Regeneration/physiology , Neurodegenerative Diseases/physiopathology , Patents as Topic
13.
Arthritis Rheumatol ; 68(2): 347-58, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26473505

ABSTRACT

OBJECTIVE: Transforming growth factor ß-activated kinase 1 (TAK1) is a key MAPKKK family protein in interleukin-1ß (IL-1ß), tumor necrosis factor (TNF), and Toll-like receptor signaling. This study was undertaken to examine the posttranslational modification of TAK1 and its therapeutic regulation in rheumatoid arthritis (RA). METHODS: The effect of TAK1, IL-1 receptor-associated kinase 1 (IRAK-1), and TNF receptor-associated factor 6 (TRAF6) inhibition was evaluated in IL-1ß-stimulated human RA synovial fibroblasts (RASFs). Western blotting, immunoprecipitation, and 20S proteasome assay were used to study the ubiquitination process in RASFs. The efficacy of epigallocatechin-3-gallate (EGCG), a potent antiinflammatory molecule, in regulating these processes in RASFs was evaluated. Molecular docking was performed to examine the interaction of EGCG with human TAK1, IRAK-1, and TRAF6. These findings were confirmed using a rat model of adjuvant-induced arthritis (AIA). RESULTS: Inhibition of TAK1, but not IRAK-1 or TRAF6, completely abrogated IL-1ß-induced IL-6 and IL-8 synthesis in RASFs. EGCG inhibited TAK1 phosphorylation at Thr(184/187) and occupied the C(174) position, an ATP-binding site, to inhibit its kinase activity. EGCG pretreatment also inhibited K(63) -linked autoubiquitination of TRAF6, a posttranslational modification essential for TAK1 autophosphorylation, by forming a stable H bond at the K(124) position on TRAF6. Furthermore, EGCG enhanced proteasome-associated deubiquitinase expression to rescue proteins from proteasomal degradation. Western blot analyses of joint homogenates from rats with AIA showed a significant increase in K(48) -linked polyubiquitination, TAK1 phosphorylation, and TRAF6 expression when compared to naive rats. Administration of EGCG (50 mg/kg/day) for 10 days ameliorated AIA in rats by reducing TAK1 phosphorylation and K(48) -linked polyubiquitination. CONCLUSION: Our findings provide a rationale for targeting TAK1 for the treatment of RA with EGCG.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Catechin/analogs & derivatives , Fibroblasts/drug effects , Interleukin-1beta/pharmacology , MAP Kinase Kinase Kinases/drug effects , TNF Receptor-Associated Factor 6/drug effects , Ubiquitination/drug effects , Animals , Blotting, Western , Catechin/pharmacology , Disease Models, Animal , Female , Fibroblasts/metabolism , Humans , Immunoprecipitation , In Vitro Techniques , Interleukin-1 Receptor-Associated Kinases/drug effects , Interleukin-1 Receptor-Associated Kinases/metabolism , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/metabolism , Lysine/metabolism , MAP Kinase Kinase Kinases/metabolism , Molecular Docking Simulation , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational/drug effects , Rats , Rats, Inbred Lew , Synovial Membrane/cytology , TNF Receptor-Associated Factor 6/metabolism
14.
J Mol Neurosci ; 57(3): 335-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26240040

ABSTRACT

The objective of this study is to explore the neuroprotective effect and mechanism of picroside II on ERK1/2-COX2 signal transduction pathway after cerebral ischemic injury in rats. Focal cerebral ischemic models were established by inserting monofilament threads into the middle cerebral artery in 200 Wistar rats. Twenty four rats were randomly selected into control group, while the other rats were randomly divided into six groups: model group, picroside group, lipopolysaccharide (LPS) with picroside group, U0126 with picroside group, LPS group, and U0126 group with each group containing three subgroups with ischemia at 6, 12, and 24 h. Neurobehavioral function in the rats was evaluated by modified neurological severity score points (mNSS) test; structure of neurons was observed using hematoxylin-eosin (HE) staining; apoptotic cells were counted using terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay; expressions of phosphorylated mitogen/extracellular signal-regulated kinase kinas1/2 (pMEK1/2), phosphorylated extracellular signal-regulated protein kinase1/2 (pERK1/2), and cyclooxygenase (COX2) in the cortex were determined using immunohistochemistry (IHC) and Western blot (WB); and real-time PCR was used to determine the level of COX2 mRNA. The neurological behavioral malfunction appeared in all rats with middle cerebral artery occlusion (MCAO). In the model group, neuron damage was extensive, while the neurobehavioral function score, apoptotic cell index, expression of pMEK1/2, pERK1/2, and COX2 and the level of COX2 mRNA increased significantly when compared to the control group. The peak COX2 mRNA level was in ischemia 12 h, prior to the peak in COX2 protein expression. In the picroside and U0126 groups, the neurological behavioral function was improved, and the number of apoptotic cells and the expression of pMEK1/2, pERK1/2, and COX2 decreased significantly when compared to the model group. In the LPS with picroside group, at ischemia 6 h neuron damage was extensive, and pMEK1/2, pERK1/2, and COX2 expression were much higher than in the model group. But at ischemia 12 and 24 h, the expression of pMEK1/2, pERK1/2, and COX2 decreased slightly, and the neurobehavioral function also improved slightly. In LPS group, neuron damage was extensive, pMEK1/2, pERK1/2, and COX2 expression was still at a high level, and COX2 mRNA peak arrived at ischemic 12 h. Picroside II downregulates COX2 expression after MCAO by inhibiting MEK-ERK1/2 in rats to protect neurons from apoptosis and inflammation.


Subject(s)
Brain Damage, Chronic/prevention & control , Cinnamates/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Iridoid Glucosides/pharmacology , Neuroprotective Agents/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain Damage, Chronic/etiology , Brain Damage, Chronic/pathology , Butadienes/pharmacology , Cerebral Cortex/pathology , Cinnamates/therapeutic use , Cyclooxygenase 2/biosynthesis , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2 Inhibitors/therapeutic use , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Infarction, Middle Cerebral Artery/pathology , Iridoid Glucosides/therapeutic use , Lipopolysaccharides/toxicity , MAP Kinase Kinase Kinases/biosynthesis , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/biosynthesis , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/genetics , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroprotective Agents/therapeutic use , Nitriles/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Wistar , Severity of Illness Index
15.
Diab Vasc Dis Res ; 12(4): 290-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25963575

ABSTRACT

Pro-inflammatory conditions induced by products of protein glycation in diabetes substantially enhance the risk of endothelial dysfunction and related vascular complications. Endothelial cell specific molecule-1 (ESM-1) or endocan has been demonstrated as a potential biomarker in cancer and sepsis. Its role in diabetes-induced pathologies remains unknown. The expression of ESM-1 gene is under cytokine regulation, indicating its role in endothelium-dependent pathological disorders. In this study, we investigated the effect of advanced glycated human serum albumin (AGE-HSA) on the production of ESM-1. We show that AGE-HSA exerts a modulating role on the expression of ESM-1 in human umbilical vein endothelial cells. It up-regulates expression of ESM-1 protein in a dose-dependent manner which correlates with its messenger RNA (mRNA) transcription. RAGE and galectin-3, both AGE receptors, show antagonistic action on its expression. While gene silencing of RAGE has down-regulatory effect, that of galectin-3 has up-regulatory effect on AGE-induced expression of ESM-1. Inhibition of MAPKKK and JNK pathways did not alter the expression. In contrast, phosphatidylinositol 3 kinase (PI3K) inhibition significantly up-regulated ESM-1 expression. In conclusion, these results suggest that AGE-induced activation of human umbilical vein endothelial cells promotes formation of endocan which is an endothelial dysfunction marker and may be related to vascular disease in diabetes.


Subject(s)
Diabetes Mellitus/physiopathology , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Glycation End Products, Advanced/pharmacology , Neoplasm Proteins/drug effects , Proteoglycans/drug effects , Serum Albumin/pharmacology , Diabetes Mellitus/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Galectin 3/drug effects , Galectin 3/metabolism , Human Umbilical Vein Endothelial Cells , Humans , JNK Mitogen-Activated Protein Kinases/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Kinase Kinases/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteoglycans/genetics , Proteoglycans/metabolism , Real-Time Polymerase Chain Reaction , Receptor for Advanced Glycation End Products/drug effects , Receptor for Advanced Glycation End Products/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Glycated Serum Albumin
16.
Neurochem Res ; 40(6): 1220-31, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25998883

ABSTRACT

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been recently shown to elicit inflammatory response in a number of cell-types. However, whether TCDD could provoke inflammation in astrocytes, the most abundant glial cells in central nervous system (CNS), remains virtually unknown. In the present study, we showed that TCDD exposure could induce evident astrocyte activation both in vivo and in vitro. Further, we found that TGF-ß-activated kinase 1 (TAK1), a critical regulator of NF-κB signaling, was rapidly phosphorylated in the process of TCDD-induced reactive astroglia. Exposure to TCDD led to rapid TAK1 and NF-κB p65 phosphorylation, as well as IKBα degradation. Moreover, blockage of TAK1 using siRNA oligos or TAK1 inhibitor 5Z-7-oxozeaenol significantly attenuated TCDD-induced astrocyte activation as well as the release of TNF-α. Finally, we showed that the conditioned medium of TCDD-treated astrocytes promoted the apoptosis of PC12 neuronal cells, which could be blocked with the pre-treatment of TAK1 inhibitor. Taken together, these findings suggested that TCDD could promote the inflammatory activation of astrocytes through modulating TAK1-NF-κB cascade, implicating that reactive astrocytes might contribute to TCDD-induced adverse effects on CNS system.


Subject(s)
Astrocytes/drug effects , Environmental Pollutants/toxicity , MAP Kinase Kinase Kinases/drug effects , NF-kappa B/drug effects , Neurons/drug effects , Polychlorinated Dibenzodioxins/toxicity , Animals , Cell Death/drug effects , Cells, Cultured , Culture Media, Conditioned , Female , I-kappa B Proteins/drug effects , I-kappa B Proteins/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , PC12 Cells , Phosphorylation , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/metabolism
17.
Inflamm Res ; 64(6): 423-31, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25917044

ABSTRACT

AIMS AND OBJECTIVE: Wogonin has multiple pharmacological effects, including anti-inflammatory effects. Here, we hypothesize that wogonin can protect intestinal barrier function in lipopolysaccharide (LPS)-induced Caco-2 cells, which is an in vitro model of intestinal inflammation. METHODS: We measured intestinal barrier function in LPS-induced Caco-2 cells by using transepithelial electrical resistance (TEER) and transport of fluorescent markers. A quantitative (q) RT-PCR and immunofluorescent staining analysis was used to detect the expression of tight junction proteins (claudin-1 and ZO-1) in LPS-induced Caco-2 cells. We measured inflammatory molecules in LPS-induced Caco-2 cells using ELISA and qRT-PCR. In addition, the expression of TLR4, MyD88 and TAK1 and their interaction, and NF-κB activity in LPS-induced Caco-2 cells were investigated by western blot analysis and immune-precipitation. RESULTS: We found that exposing Caco-2 cells to wogonin (10 and 50 µM for 24 h) attenuated the LPS-induced changes in TEER and transport of fluorescent markers. In addition, wogonin suppressed LPS-induced down-regulation of tight junction proteins (claudin-1 and ZO-1). Furthermore, LPS-induced up-regulation of inflammatory mediators, including interleukin (IL)-1ß, IL-6 and IL-8, cyclooxygenase-2 (COX-2), inducible nitric oxide synthases (iNOS) were reduced after being pre-treated with wogonin. Moreover, wogonin not only inhibited the expression of TLR4, MyD88 and TAK1 and the interaction between these molecules, but also reduced NF-κB translocation to nucleus and its DNA-binding activity in LPS-induced Caco-2 cells. CONCLUSION: Our results suggested that pre-treatment with wogonin could attenuate the TLR4-mediated inflammatory response and maintain intestinal barrier function in LPS-induced Caco-2 cells, thus might be a potential therapy for treating IBD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Flavanones/pharmacology , Intestines/drug effects , MAP Kinase Kinase Kinases/drug effects , Myeloid Differentiation Factor 88/drug effects , NF-kappa B/drug effects , Signal Transduction/drug effects , Toll-Like Receptor 4/drug effects , Biological Transport, Active/drug effects , Caco-2 Cells , Claudin-1/biosynthesis , Claudin-1/genetics , Electric Impedance , Humans , Inflammation Mediators/metabolism , Interleukins/metabolism
18.
Br J Pharmacol ; 172(11): 2892-904, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25625840

ABSTRACT

BACKGROUND AND PURPOSE: Tolerance induced by morphine and other opiates remains a major unresolved problem in the clinical management of pain. There is now good evidence for the importance of MAPKs in morphine-induced antinociceptive tolerance. A member of the MAPK kinase kinase family, TGF-ß activated kinase 1 (TAK1) is the common upstream kinase of MAPKs. Here, we have assessed the involvement of TAK1 in the development of tolerance to morphine-induced analgesia. EXPERIMENTAL APPROACH: The effects of an antagonist of TAK1 on morphine tolerance were investigated in vivo using the Randall-Selitto test, and the mechanism was investigated using Western blot and immunohistochemistry. The expression of TAK1 after chronic morphine exposure was also evaluated in vitro by immunohistochemistry. KEY RESULTS: Chronic intrathecal morphine exposure up-regulated protein levels and phosphorylation of spinal TAK1. TAK1 immunoreactivity was co-localized with the neuronal marker NeuN. Intrathecal administration of 5Z-7-oxozeaenol (OZ), a selective TAK1 inhibitor, attenuated the loss of morphine analgesic potency and morphine-induced TAK1 up-regulation. Furthermore, OZ decreased the up-regulated expression of spinal p38 and JNK after repeated morphine exposure. In vitro studies demonstrated that sustained morphine treatment induced TAK1 up-regulation, which was reversed by co-administration of OZ. A bolus injection of OZ showed some reversal of established morphine antinociceptive tolerance. CONCLUSIONS AND IMPLICATIONS: TAK1 played a pivotal role in the development of morphine-induced antinociceptive tolerance. Modulation of TAK1 activation by the selective inhibitor OZ in the lumbar spinal cord may prove to be an attractive adjuvant therapy to attenuate such tolerance.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance , MAP Kinase Kinase Kinases/drug effects , Morphine/pharmacology , Nociception/drug effects , Spinal Cord/drug effects , Analgesia , Animals , Immunohistochemistry , Infusions, Spinal , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Male , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Up-Regulation , Zearalenone/analogs & derivatives , Zearalenone/pharmacology
19.
Pharmazie ; 69(3): 229-33, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24716415

ABSTRACT

BACKGROUND: Curcumin is a multi-targeted anti-cancer agent. However, there are few studies on its anti-leukemia activity in human acute monocytic leukemia. Here, we study the effect and mechanisms of curcumin on acute monocytic leukemia. METHODS: The acute monocytic leukemia cell line THP-1 was used as in vitro cell model to explore the anti-leukemia effects and mechanisms of curcumin. Cell proliferation was measured by MTT assay, cell apoptosis bodies were observed using a light microscope, cell apoptosis rate was evaluated by flow cytometry, and the expression alterations of growth-sinaling proteins were detected by Western blotting. RESULTS: Curcumin inhibited cell proliferation and induced cell apoptosis in time- and dose- dependent manner in THP-1 cells. Curcumin significantly inhibited the activations of AKT/mTOR and RAF/MEK/ERK signaling pathways simultaneously. CONCLUSION: This study demonstrates that curcumin inhibits proliferation and induces apoptosis in THP-1 cells via inhibiting the activations of AKT/mTOR and RAF/MEK/ERK signaling pathways simultaneously. Our data suggest that curcumin is a promising anti-tumor agent in acute monocytic leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Curcumin/pharmacology , Leukemia, Myeloid, Acute/drug therapy , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Signaling System/drug effects , Oncogene Protein v-akt/drug effects , TOR Serine-Threonine Kinases/drug effects , raf Kinases/drug effects , Annexin A5 , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Coloring Agents , Enzyme Activation/drug effects , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Leukemia, Myeloid, Acute/pathology , Tetrazolium Salts , Thiazoles
20.
J Dent Res ; 92(1): 51-7, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23114032

ABSTRACT

The discovery that dental pulp stem cells are capable of differentiating into endothelial cells raises the exciting possibility that these cells can be a single source of odontoblasts and vascular networks in dental tissue engineering. The purpose of this study was to begin to define signaling pathways that regulate endothelial differentiation of SHED. Stem cells from exfoliated deciduous teeth (SHED) exposed to endothelial growth medium (EGM-2MV) supplemented with vascular endothelial growth factor (VEGF) differentiated into VEGFR2-positive and CD31-positive endothelial cells in vitro. In vivo, VEGFR1-silenced SHED seeded in tooth slice/ scaffolds and transplanted into immunodeficient mice showed a reduction in human CD31-positive blood vessels as compared with controls (p = 0.02). Exposure of SHED to EGM2-MV supplemented with VEGF induced potent activation of ERK and Akt signaling, while it inhibited phosphorylation of STAT3. Notably, genetic (MEK1 silencing) or chemical (U0126) inhibition of ERK signaling restored constitutive STAT3 phosphorylation and inhibited the differentiation of SHED into endothelial cells. Collectively, analysis of these data unveiled the VEGF/MEK1/ERK signaling pathway as a key regulator of the endothelial differentiation of dental pulp stem cells.


Subject(s)
Dental Pulp/cytology , Endothelial Cells/physiology , MAP Kinase Signaling System/physiology , Stem Cells/physiology , Tooth, Deciduous/cytology , Animals , Blood Vessels/immunology , Butadienes/pharmacology , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Differentiation/physiology , Culture Media , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Silencing/physiology , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase Kinases/drug effects , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Mice, SCID , Nitriles/pharmacology , Oncogene Protein v-akt/drug effects , Phosphorylation , Platelet Endothelial Cell Adhesion Molecule-1/analysis , STAT3 Transcription Factor/antagonists & inhibitors , Tissue Scaffolds , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-2/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...