Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Toxicol Appl Pharmacol ; 483: 116818, 2024 02.
Article in English | MEDLINE | ID: mdl-38215994

ABSTRACT

The recurrence and metastasis in breast cancer within 3 years after the chemotherapies or surgery leads to poor prognosis with approximately 1-year overall survival. Large-scale scanning research studies have shown that taking lipid-lowering drugs may assist to reduce the risk of death from many cancers, since cholesterol in lipid rafts are essential for maintain integral membrane structure and functional signaling regulation. In this study, we examined five lipid-lowering drugs: swertiamarin, gemfibrozil, clofibrate, bezafibrate, and fenofibrate in triple-negative breast cancer, which is the most migration-prone subtype. Using human and murine triple-negative breast cancer cell lines (Hs 578 t and 4 T1), we found that fenofibrate displays the highest potential in inhibiting the colony formation, wound healing, and transwell migration. We further discovered that fenofibrate reduces the activity of pro-metastatic enzymes, matrix metalloproteinases (MMP)-9 and MMP-2. In addition, epithelial markers including E-cadherin and Zonula occludens-1 are increased, whereas mesenchymal markers including Snail, Twist and α-smooth muscle actin are attenuated. Furthermore, we found that fenofibrate downregulates ubiquitin-dependent GDF-15 degradation, which leads to enhanced GDF-15 expression that inhibits cell migration. Besides, nuclear translocation of FOXO1 is also upregulated by fenofibrate, which may responsible for GDF-15 expression. In summary, fenofibrate with anti-cancer ability hinders TNBC from migration and invasion, and may be beneficial to repurposing use of fenofibrate.


Subject(s)
Fenofibrate , Triple Negative Breast Neoplasms , Animals , Humans , Mice , Triple Negative Breast Neoplasms/metabolism , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Growth Differentiation Factor 15/pharmacology , Growth Differentiation Factor 15/therapeutic use , Cell Line, Tumor , Cell Movement , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/therapeutic use , Epithelial-Mesenchymal Transition , Lipids , Cell Proliferation
2.
Front Pharmacol ; 14: 1292137, 2023.
Article in English | MEDLINE | ID: mdl-38111379

ABSTRACT

Background: Pulmonary fibrosis features in damaged pulmonary structure or over-produced extracellular matrix and impaired lung function, leading to respiratory failure and eventually death. Fibrotic lungs are characterized by the secretion of pro-fibrotic factors, transformation of fibroblasts to myofibroblasts, and accumulation of matrix proteins. Hypothesis/purpose: Imperatorin shows anti-inflammatory effects on alveolar macrophages against acute lung injury. We attempt to evaluate the properties of imperatorin on the basis of fibroblasts. Methods: In in vitro, zymosan was introduced to provoke pro-fibrotic responses in NIH/3T3 or MRC-5 pulmonary fibroblasts. Imperatorin was given for examining its effects against fibrosis. The mice were stimulated by bleomycin, and imperatorin was administered to evaluate the prophylactic potential in vivo. Results: The upregulated expression of connective tissue growth factor (CTGF), α-smooth muscle actin (α-SMA), and collagen protein due to zymosan introduction was decreased by imperatorin in fibroblasts. Zymosan induced the activity of transglutaminase 2 (TGase2) and lysyl oxidase (LOX), which was also inhibited by the administration of imperatorin. Imperatorin alone enhanced sirtuin 1 (SIRT1) activity and growth differentiation factor 15 (GDF15) secretion in fibroblasts via LKB1/AMPK/CREB pathways. In addition, GDF15 exerted a beneficial effect by reducing the protein expression of CTGF, α-SMA, and collagen and the activities of TGase and LOX. Moreover, orally administered imperatorin showed prophylactic effects on bleomycin-induced pulmonary fibrosis in mice. Conclusion: Imperatorin reduces fibrotic marker expression in fibroblasts and also increases GDF15 secretion via the LKB1/AMPK/CREB pathway, attenuating pro-fibrotic responses in vitro. Imperatorin also alleviates pulmonary fibrosis induced by bleomycin in vivo.

3.
Oncogenesis ; 11(1): 48, 2022 Aug 13.
Article in English | MEDLINE | ID: mdl-35963873

ABSTRACT

Migration and metastasis commonly happen to triple-negative breast cancer (TNBC) patients with advanced diseases. In many studies, it has been suggested that epithelial-mesenchymal transition (EMT) is one of the key mechanisms triggering cancer metastasis. Accumulating evidence has proven that calcium channel blockers mediate cell motility. Therefore, we attempt to investigate the effects of diltiazem, which has been selected from several FDA-approved clinical calcium channel blockers, on EMT in TNBC. By using both mouse and human TNBC cell lines, we found that diltiazem decreases colony formation and cell migration in breast cancer cells. The expression of epithelial markers such as E-cadherin and ZO-1 were increased dose-dependently by diltiazem, while mesenchymal markers such as Snail and Twist were decreased. In addition, we found that the expression of growth differentiation factor-15 (GDF-15) was also increased by diltiazem. Administering recombinant GDF-15 also reverses EMT, inhibits colony formation and migration in breast cancer cells. Moreover, treatment with diltiazem in tumor-bearing mice also decreases cancer metastasis and nodule formation, with more GDF-15 expression in diltiazem-treated mice than saline-treated mice, respectively. These findings suggest that diltiazem regulates EMT and cell motility through elevating GDF-15 expression in breast cancers in vitro and in vivo.

4.
Int J Comput Assist Radiol Surg ; 17(4): 661-671, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35257285

ABSTRACT

PURPOSE: Non-contrast computed tomography (NCCT) is a first-line imaging technique for determining treatment options for acute ischemic stroke (AIS). However, its poor contrast and signal-to-noise ratio limit the diagnosis accuracy for radiologists, and automated AIS lesion segmentation using NCCT also remains a challenge. In this paper, we propose R2U-RNet, a novel model for AIS lesion segmentation using NCCT. METHODS: We used an in-house retrospective NCCT dataset with 261 AIS patients with manual lesion segmentation using follow-up diffusion-weighted images. R2U-RNet is based on an R2U-Net backbone with a novel residual refinement unit. Each input image contains two image channels from separate preprocessing procedures. The proposed model incorporates multiscale focal loss to mitigate the class imbalance problem and to leverage the importance of different levels of details. A proposed noisy-label training scheme is utilized to account for uncertainties in the manual annotations. RESULTS: The proposed model outperformed several iconic segmentation models in AIS lesion segmentation using NCCT, and our ablation study demonstrated the efficacy of the proposed model. Statistical analysis of segmentation performance revealed significant effects of regional stroke occurrence and side of the stroke, suggesting the importance of region-specific information for automated segmentation, and the potential influence of the hemispheric difference in clinical data. CONCLUSION: This study demonstrated the potentials of R2U-RNet model for automated NCCT AIS lesion segmentation. The proposed model can serve as a tool for accelerating AIS diagnoses and improving the treatment quality of AIS patients.


Subject(s)
Ischemic Stroke , Stroke , Humans , Ischemic Stroke/diagnostic imaging , Retrospective Studies , Signal-To-Noise Ratio , Stroke/diagnostic imaging , Tomography, X-Ray Computed
5.
Food Chem Toxicol ; 159: 112761, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34890758

ABSTRACT

Pulmonary inflammation involves complex immune responses in which alveolar macrophages release pro-inflammatory proteins and cytokines. Cardamonin is a spice component that exerts anti-inflammatory and anti-oxidative properties against pulmonary inflammation. Herein, the aim of this research is to investigate the effects of cardamonin on pulmonary inflammation and its mechanism. Pulmonary inflammation in mice was induced by intratracheal administration of PMA. PMA-stimulated acute fibrosis, pulmonary edema, and inflammatory responses were ameliorated by oral administration of cardamonin in vivo. In MH-S alveolar macrophages, PMA-induced pro-inflammatory responses, including iNOS, COX-2, MMP-9 and cytokines expressions were reduced by cardamonin. The anti-oxidative Nrf2/HO-1 axis was also provoked by cardamonin in MH-S alveolar macrophages. In addition, MMP-9 expression induced by PMA is also decreased by the down-stream metabolites of HO-1, indicating that HO-1 expression partially contributes to the anti-inflammatory effect exerted by cardamonin. In this study, cardamonin demonstrates anti-inflammatory and anti-oxidative effects on PMA-induced pulmonary inflammation and activating Nrf2/HO-1 axis in alveolar macrophages. Cardamonin also ameliorates pulmonary inflammation, rapid fibrosis in vivo, suggesting powerful health benefits.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chalcones/pharmacology , Macrophages, Alveolar/drug effects , Pneumonia/metabolism , Tetradecanoylphorbol Acetate/toxicity , Animals , Heme Oxygenase-1 , Lung/drug effects , Lung/pathology , Membrane Proteins , Mice , NF-E2-Related Factor 2 , Pneumonia/pathology
6.
Front Pharmacol ; 12: 710978, 2021.
Article in English | MEDLINE | ID: mdl-34483918

ABSTRACT

Background: Metastasis represents an advanced stage of cancers, and matrix metalloproteinases are critical regulators. Calcium signal is crucial for appropriate cell behaviors. The efficacy and effects of calcium channel blockers in treating cancers are individually differ from each other. Here, we attempt to investigate the effects of nicardipine, a FDA-approved calcium channel blocker, in advanced breast cancers. Methods: We analyzed the influence of nicardipine on the colony-forming ability of triple negative breast cancer cell lines. Using cell culture inserts, cell migration was also examined. The expression of regulatory proteins was evaluated by real-time PCR, Western blot, and ELISA. Results: We have confirmed that nicardipine inhibits the breast cancer cells migration and colony formation. In addition, we also revealed that nicardipine increases the Nrf2 and HO-1 expression. The inhibition of HO-1 abrogates nicardipine-reduced matrix metalloproteinase-9 expression. Moreover, the end products of HO-1, namely, CO, Fe2+, and biliverdin (will converted to bilirubin), also decreases the expression of matrix metalloproteinase-9. Conclusion: These findings suggest that nicardipine-mediated matrix metalloproteinase-9 reduction is regulated by Nrf2/HO-1 axis and its catalytic end products. Therefore, nicardipine may be a potential candidate for repurposing against advanced breast cancers.

7.
Ther Adv Med Oncol ; 11: 1758835919846806, 2019.
Article in English | MEDLINE | ID: mdl-31205504

ABSTRACT

BACKGROUND: Leptin is considered a tumorigenic adipokine, suggested to promote tumorigenesis and progression in many cancers. On the other hand, intercellular adhesion molecule-1 (ICAM-1) shows altered expression in a variety of benign and malignant diseases. Histologically, ICAM-1 expression is reported as proportional to cancer stage and considered as a potential diagnosis biomarker. The altered expressions of ICAM-1 and its soluble form in malignant diseases have gained interests in recent years. MATERIAL AND METHODS: The expression of ICAM-1 and its regulatory signaling were examined by Western blot or flow cytometry. The effect of soluble ICAM-1 on osteoclast formation was investigated by tartrate-resistance acid phosphatase staining of RAW cells and tumor-induced osteolysis in vivo. RESULTS: In our study, we found that leptin enhanced soluble ICAM-1 production but not surface ICAM-1 expression in lung and breast cancer cells, and this effect was regulated through leptin receptor (ObR), while silencing ObR abrogated leptin-induced soluble ICAM-1 expression. In addition, we revealed that leptin administration provoked the JAK1/2, STAT3, FAK, ERK, and GSK3αß signaling cascade, leading to the elevation of ICAM-1 expression. Moreover, soluble ICAM-1 secreted by leptin-stimulated cancer cells synergize with the receptor activator of nuclear factor kappa-B ligand (RANKL) in inducing osteoclast formation. Soluble ICAM also enhanced tumor-induced osteolysis in vivo. CONCLUSION: These findings suggest that soluble ICAM-1 produced under leptin treatment enhances osteoclast formation and is involved in tumor-induced osteolysis.Leptin plays an important role in physiology in health and diseases. Leptin affects immune responses that may induce inflammation and carcinogenesis. Leptin is also considered as a tumorigenic adipokine suggested to promote tumorigenesis and progression in many cancers. On the other hand, intercellular adhesion molecule-1 (ICAM-1) shows altered expression in a variety of benign and malignant diseases. Histologically, ICAM-1 expression is reported to be proportional to cancer stage and considered as a potential diagnosis biomarker. It has been reported that soluble ICAM-1 allows tumor cells to escape from immune recognition and stimulates angiogenesis and tumor growth. The altered expressions of ICAM-1 and its soluble form in malignant diseases have gained interests in recent years. In our study, we found that leptin enhanced soluble ICAM-1 production but not surface ICAM-1 expression in lung and breast cancer cells, and this effect was regulated through leptin receptor (ObR), while silencing ObR abrogated leptin-induced soluble ICAM-1 expression. In addition, we revealed that leptin administration provoked the JAK1/2, STAT3, FAK, ERK, and GSK3αß signaling cascade, leading to the elevation of ICAM-1 expression. Moreover, soluble ICAM-1 secreted by leptin-stimulated cancer cells synergize with receptor activator of nuclear factor-kappa B ligand in inducing osteoclast formation. Soluble ICAM also enhanced tumor-induced osteolysis in vivo. These findings suggest that soluble ICAM-1 produced under leptin treatment is possibly involved in lung and breast cancer bone metastasis.

8.
Int J Mol Sci ; 20(1)2018 Dec 25.
Article in English | MEDLINE | ID: mdl-30585203

ABSTRACT

OBJECTIVE: Intervertebral disc (IVD) degeneration and disc herniation are major causes of lower back pain, which involve the presence of inflammatory mediators and tissue invasion by immune cells. Intercellular adhesion molecule 1 (ICAM1, also termed CD54) is an adhesion molecule that mediates cell-cell interactions, particularly between immune cells and target tissue. The aim of this study was to examine the intracellular signaling pathways involved in inflammatory stimuli-induced ICAM1 expression in human anulus fibrosus (AF) cells. METHODS: Quantitative reverse transcription-polymerase chain reaction (qPCR), western blotting, and flow cytometry were performed to dissect the roles of different signaling pathways in inflammatory stimuli-mediated ICAM1 expression. RESULTS: Using qPCR and western blot analyses, a significant increase in ICAM1 expression was observed in AF cells after stimulation of lipopolysaccharide (LPS) plus interferon-gamma (IFNγ) in a time-dependent manner. Flow cytometry revealed ICAM1 upregulation on the surface of AF cells. Importantly, LPS plus IFNγ treatment also significantly promoted Chemokine ligand (CCL)2 expression, but not CCL3. The enhanced ICAM1 expression was abolished after incubation with antibody against CCL2. In AF cells, treatment with LPS plus IFNγ activated the FAK/ERK/GSK3 signaling pathways, promoted a time-dependent increase in PKCδ phosphorylation, and promoted PKCδ translocation to the nucleus. Treatment with the pharmacological PKCδ inhibitor; rottlerin, effectively blocked the enhanced productions of ICAM1 and CCL2. CONCLUSIONS: Inflammatory stimuli in AF cells are part of a specific pathophysiology in IVD degeneration and disc herniation that modulates CCL2/ICAM1 activation through the FAK/ERK/GSK3 and PKCδ signaling pathways in AF cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesion Kinase 1/metabolism , Glycogen Synthase Kinase 3/metabolism , Intercellular Adhesion Molecule-1/metabolism , Protein Kinase C-delta/metabolism , Acetophenones/pharmacology , Annulus Fibrosus/cytology , Annulus Fibrosus/metabolism , Benzopyrans/pharmacology , Chemokine CCL2/metabolism , Humans , Interferon-gamma/pharmacology , Janus Kinase 2/metabolism , Lipopolysaccharides/pharmacology , Phosphorylation/drug effects , Protein Kinase C-delta/antagonists & inhibitors , Signal Transduction/drug effects , Up-Regulation/drug effects
9.
Food Chem Toxicol ; 120: 528-535, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30076913

ABSTRACT

Metastasis is commonly seen in advanced stage of cancers, and matrix metalloproteinases (MMPs) are commonly up-regulated and have been identified as critical regulators. In this present study, a flavonoid, fisetin, which can be found in diverse foods, is investigated for its ability to inhibit cell motility, and the underlying mechanism is also studied in breast cancer cells (4T1 and JC cells). We have revealed that fisetin increased HO-1 mRNA and protein expressions. Besides, fisetin also elevated Nrf2 expression in nuclear fraction. By silencing Nrf2, fisetin-induced HO-1 expression was abrogated, suggested that HO-1 expression was mediated by up-regulation of the transcription factor Nrf2. In addition, we also found that fisetin decreased MMP-2 and MMP-9 enzyme activity and gene expression in both protein and mRNA levels. Moreover, by administration of HO-1 inhibitors, tin protoporphyrin and zinc protoporphyrin, fisetin-reduced MMP-2 and MMP-9 expressions were reversed. Furthermore, transfection of siRNA against HO-1 and Nrf2 also abolished MMP-2 and MMP-9 reduction exerted by fisetin. These findings suggest that fisetin-mediated MMP-2 and MMP-9 reduction is regulated by HO-1 through Nrf2. Therefore, fisetin may be useful as a potential therapeutic agent for the treatment of metastatic breast cancer.


Subject(s)
Flavonoids/pharmacology , Heme Oxygenase-1/biosynthesis , Matrix Metalloproteinases/biosynthesis , Triple Negative Breast Neoplasms/enzymology , Triple Negative Breast Neoplasms/pathology , Cell Line, Tumor , Female , Flavonols , Humans , NF-E2-Related Factor 2/physiology , Neoplasm Metastasis
10.
Mol Neurobiol ; 55(9): 7487-7499, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29427085

ABSTRACT

Inhibition of microglial over-activation is an important strategy to counter balance neurodegenerative progression. We previously demonstrated that the adenosine monophosphate-activated protein kinase (AMPK) may be a therapeutic target in mediating anti-neuroinflammatory responses in microglia. Brain-derived neurotrophic factor (BDNF) is one of the major neurotrophic factors produced by astrocytes to maintain the development and survival of neurons in the brain, and have recently been shown to modulate homeostasis of neuroinflammation. Therefore, the present study focused on BDNF-mediated neuroinflammatory responses and may provide an endogenous regulation of neuroinflammation. Among the tested neuroinflammation, epigallocatechin gallate (EGCG) and minocycline exerted BDNF upregulation to inhibit COX-2 and proinflammatory mediator expressions. Furthermore, both EGCG and minocycline upregulated BDNF expression in microglia through AMPK signaling. In addition, minocycline and EGCG also increased expressions of erythropoietin (EPO) and sonic hedgehog (Shh). In the endogenous modulation of neuroinflammation, astrocyte-conditioned medium (AgCM) also decreased the expression of COX-2 and upregulated BDNF expression in microglia. The anti-inflammatory effects of BDNF were mediated through EPO/Shh in microglia. Our results indicated that the BDNF-EPO-Shh novel-signaling pathway underlies the regulation of inflammatory responses and may be regarded as a potential therapeutic target in neurodegenerative diseases. This study also reveals a better understanding of an endogenous crosstalk between astrocytes and microglia to regulate anti-inflammatory actions, which could provide a novel strategy for the treatment of neuroinflammation and neurodegenerative diseases.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Inflammation/pathology , Microglia/metabolism , Signal Transduction , Animals , Anti-Inflammatory Agents/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line , Culture Media, Conditioned/pharmacology , Cyclooxygenase 2/metabolism , Erythropoietin/metabolism , Hedgehog Proteins/metabolism , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides , Mice , Microglia/drug effects , Microglia/pathology , Minocycline/pharmacology , Models, Biological , Neuroprotective Agents/pharmacology
11.
Toxicol Appl Pharmacol ; 338: 182-190, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29180066

ABSTRACT

Connexins are widely supported as tumor suppressors due to their downregulation in cancers, nevertheless, more recent evidence suggests roles for connexins in facilitating tumor progression in later stages, including metastasis. One of the key factors regulating the expression, modification, stability, and localization of connexins is hormone receptors in hormone-dependent cancers. It is reasonable to consider that hormones/hormone receptors may modulate connexins expression and play critical roles in the cellular control of connexins during breast cancer progression. In estrogen receptor (ER)-positive breast cancers, tamoxifen and fulvestrant are widely used therapeutic agents and are considered to alter ER signaling. In this present study, we investigated the effects of fulvestrant and tamoxifen in Cx43 expression, and we also explored the role of Cx43 in ER-positive breast cancer migration and the relationship between Cx43 and ER. The involvement of estrogen/ER in Cx43 modulation was further verified by administering tyrosine kinase inhibitors and chemotherapeutic agents. We found that inhibition of ER promoted the binding of E3 ligase Nedd4 to Cx43, leading to Cx43 ubiquitination. Furthermore, inhibition of ER by fulvestrant and tamoxifen phosphorylated p38 MAPK, and inhibition of Rac, MKK3/6, and p38 reversed fulvestrant-reduced Cx43 expression. These findings suggest that Cx43 expression which may positively regulate cell migration is ER-dependent in ER-positive breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , Connexin 43/physiology , Estrogen Antagonists/pharmacology , Breast Neoplasms/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Connexin 43/analysis , Female , Humans , Nedd4 Ubiquitin Protein Ligases/metabolism , Receptors, Estrogen/physiology , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , p38 Mitogen-Activated Protein Kinases/physiology
12.
Sci Rep ; 7(1): 8578, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819180

ABSTRACT

We found that the coagulation and cytokine pathways were important mechanisms involve in the degeneration of intervertebral discs (IVD) using a microarray approach to analyze gene expression in different grades of specimens. Furthermore, using a cytokine/chemokine array, a significant increase in CXCL8 expression was observed in human nucleus pulposus (NP) cells after thrombin treatment. The enhancement of CXCL8 expression by thrombin was activated by the PAR1 receptor. Importantly, analysis of degenerated human NP tissue samples showed that EGFR expression positively correlated with the grade of tissue degeneration. In NP cells, thrombin caused an increase in phosphorylation of the EGFR at the Tyr1068, and treatment with the pharmacological EGFR inhibitor, AG1473 effectively blocked thrombin-enhanced CXCL8 production. Surprisingly, inhibition of STAT3 for 24 h decreased expression of EGFR. Treatment with thrombin also increased Akt and GSK3α/ß activation; this activation was also blocked by EGFR inhibitor. Although c-Src, ERK, and FAK were activated by thrombin, only c-Src and ERK were involved in the STAT3/CXCL8 induction. Our findings indicate that stimulation of an inflammatory response in NP cells by thrombin is part of a specific pathophysiology that modulates the EGFR activation through activation of Src/ERK/STAT3 signaling.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , Nucleus Pulposus/drug effects , Thrombin/pharmacology , Adult , Aged , Cells, Cultured , Cytokines/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression/drug effects , Gene Expression Profiling , Humans , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Middle Aged , Nucleus Pulposus/cytology , Nucleus Pulposus/metabolism , Signal Transduction/drug effects , Young Adult
13.
Molecules ; 21(5)2016 May 05.
Article in English | MEDLINE | ID: mdl-27164068

ABSTRACT

Coronary artery disease (CAD) is the most common cause of heart attack and the leading cause of mortality in the world. It is associated with mitochondrial dysfunction and increased level of reactive oxygen species production. According to the Ottawa Heart Genomics Study genome-wide association study, a recent research identified that Q688 spastic paraplegia 7 (SPG7) variant is associated with CAD as it bypasses the regulation of tyrosine phosphorylation of AFG3L2 and enhances the processing and maturation of SPG7 protein. This study aims to identify potential compounds isolated from Traditional Chinese Medicines (TCMs) as potential lead compounds for paraplegin (SPG7) inhibitors. For the crystallographic structure of paraplegin, the disordered disposition of key amino acids in the binding site was predicted using the PONDR-Fit protocol before virtual screening. The TCM compounds saussureamine C and 3-(2-carboxyphenyl)-4(3H)-quinazolinone, have potential binding affinities with stable H-bonds and hydrophobic contacts with key residues of paraplegin. A molecular dynamics simulation was performed to validate the stability of the interactions between each candidate and paraplegin under dynamic conditions. Hence, we propose these compounds as potential candidates as lead drug from the compounds isolated from TCM for further study in drug development process with paraplegin protein for coronary artery disease.


Subject(s)
Asparagine/analogs & derivatives , Coronary Artery Disease/genetics , Drugs, Chinese Herbal/chemistry , Enzyme Inhibitors/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Quinazolinones/pharmacology , ATPases Associated with Diverse Cellular Activities , Asparagine/chemistry , Asparagine/pharmacology , Binding Sites , Computer Simulation , Coronary Artery Disease/enzymology , Crystallography, X-Ray , Drugs, Chinese Herbal/pharmacology , Enzyme Inhibitors/chemistry , Humans , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Mutation , Quinazolinones/chemistry , Structure-Activity Relationship
14.
J Mol Model ; 22(1): 32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26757915

ABSTRACT

Decreasing iron uptake and increasing iron efflux may result in cell death by oxidative inactivation of vital enzymes. Applying the dual function of neutrophil gelatinase-associated lipocalin (NGAL) could achieve the goal of iron depletion in the cancer cells. Tyr106, Lys125 or Lys134 was the key binding site for NGAL protein to sequester iron-chelating siderophores. In this study, we employed all bioactive peptides in peptide databank to dock with the siderophore-binding sites of NGAL protein by virtual screening. In addition, we performed molecular dynamics (MD) simulation to observe the molecular character and structural variation of ligand-protein interaction. Glu-Glu-Lys-Glu (EEKE), Glu-Glu-Asp-Cys-Lys (EEDCK), and Gly-Glu-Glu-Cys-Asp (GEECD) were selected preliminarily by rigorous scoring functions for further investigation. GEECD was excluded due to higher binding total energy than the others. Moreover, we also excluded EEKE due to larger influence to the stability of binding residues by the information of root mean square fluctuation (RMSF) and principal component analysis (PCA). Thus, we suggested that EEDCK was the potential bioactive peptide which had been proved to inhibit malignant cells for targeted cancer therapy. Graphical Abstract Perspective drug design of occupying the siderophore-binding sites of NGAL outside the cell temporarily by a potential short peptide until NGAL enters into the cell, and releasing the siderophore-binding sites inside the cell.


Subject(s)
Acute-Phase Proteins/chemistry , Databases, Protein , Iron/chemistry , Lipocalins/chemistry , Molecular Docking Simulation , Peptides/chemistry , Proto-Oncogene Proteins/chemistry , Acute-Phase Proteins/therapeutic use , Humans , Lipocalin-2 , Lipocalins/therapeutic use , Neoplasms/drug therapy , Peptides/therapeutic use , Proto-Oncogene Proteins/therapeutic use
15.
Mol Neurobiol ; 53(9): 6218-6227, 2016 11.
Article in English | MEDLINE | ID: mdl-26558633

ABSTRACT

The expression of matrix metalloproteinase-13 (MMP-13) has been shown to be elevated in some pathophysiological conditions and is involved in the degradation of extracellular matrix in astrocytes. In current study, the function of MMP-13 was further investigated. The conditioned medium (CM) collected from activated microglia increased interleukin (IL)-18 production and enhanced MMP-13 expression in astrocytes. Furthermore, treatment with recombinant IL-18 increased MMP-13 protein and mRNA levels in astrocytes. Recombinant IL-18 stimulation also increased the enzymatic activity of MMP-13 and the migratory activity of astrocytes, while administration of MMP-13 or pan-MMP inhibitors antagonized IL-18-induced migratory activity of astrocytes. In addition, administration of recombinant IL-18 to astrocytes led to the phosphorylation of JNK, Akt, or PKCδ, and treatment of astrocytes with JNK, PI3 kinase/Akt, or PKCδ inhibitors significantly decreased the IL-18-induced migratory activity. Taken together, the results suggest that IL-18-induced MMP-13 expression in astrocytes is regulated by JNK, PI3 kinase/Akt, and PKCδ signaling pathways. These findings also indicate that IL-18 is an important regulator leading to MMP-13 expression and cell migration in astrocytes.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Brain/cytology , Cell Movement , Interleukin-18/metabolism , Matrix Metalloproteinase 13/metabolism , Animals , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase C-delta/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Transcription Factor AP-1/metabolism , Up-Regulation
16.
Mol Neurobiol ; 53(7): 4497-508, 2016 09.
Article in English | MEDLINE | ID: mdl-26255181

ABSTRACT

Cobalt protoporphyrin (CoPP) is a potent HO-1 inducer and generally known to be an antioxidant in various cell types. Little is known about the CoPP-induced cyclooxygenase-2 (COX-2) expression and its downstream signaling in microglial cells. In current study, CoPP caused concentration- and time-dependent increases in COX-2 expression in microglial cells. Furthermore, activation of apoptosis signal-regulating kinase (ASK) 1/MAP kinase involved in CoPP-induced COX-2 expression in microglia. CoPP also induced P2X7 receptor activation, and treatment of P2X7 inhibitors effectively reduced CoPP-induced COX-2 expression. Protein inhibitor of activated STAT (PIAS) 1 is reported to be involved in modulating anti-inflammatory response through negative regulation of transcription factors. Interestingly, treatment with CoPP markedly induced PIAS1 degradation which is regulated by PI3K, Akt, and glycogen synthase kinase 3α/ß (GSK3α/ß) signaling pathways. These results suggest that CoPP induces COX-2 expression through activating P2X7 receptors and ASK1/MAP kinases as well as PIAS1 degradation signaling pathways. Our study provides a new insight into the regulatory effect of CoPP on neuroinflammation in microglial cells.


Subject(s)
Cyclooxygenase 2/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Protoporphyrins/pharmacology , Up-Regulation/drug effects , Animals , Cell Line , Down-Regulation/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Mice , Microglia/drug effects , Microglia/metabolism , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Purinergic P2X7/metabolism , Signal Transduction/drug effects
17.
Mol Neurobiol ; 53(2): 1080-1091, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25579382

ABSTRACT

Accumulating evidence suggests that neuroinflammation is closely associated with the pathogenesis of neurodegenerative disorders such as Parkinson's disease and Alzheimer's disease. The hallmark of neuroinflammation is considered to be microglial activation in the central nervous system (CNS). Activated microglia release pro-inflammatory cytokines which cause neuroinflammation and progressive neuronal cell death. Therefore, inhibition of microglial activation is considered an important strategy in the development of neuroprotective strategy. Naringenin, a flavonoid found in citrus fruits and tomatoes, has been reported to have anti-oxidant, anti-cancer, and anti-inflammatory properties. However, the mechanism of its beneficial anti-inflammatory effects in the CNS is poorly understood. In this study, we demonstrated that naringenin inhibites the release of nitric oxide (NO), the expression of inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2), as well as pro-inflammatory cytokines in microglial cells. Treatment of naringenin also induced suppressors of cytokine signaling (SOCS)-3 expression in microglia. The SOCS-3 expression and anti-inflammatory effects of naringenin were found to be regulated by adenosine monophosphate-activated protein kinase α (AMPKα) and protein kinase C δ (PKCδ). Besides, naringenin exerted protective property against neurotoxicity caused by LPS-induced microglial activation. Our findings suggest that naringenin-inhibited iNOS and COX-2 expression is mediated by SOCS-3 activation through AMPKα and PKCδ signaling pathways. In a mouse model, naringenin also showed significant protective effects on microglial activation and improved motor coordination function as well. Therefore, naringenin that involves in anti-neuroinflammatory responses and neuroprotection might be a potential agent for treatment of inflammation-associated disorders.


Subject(s)
Flavanones/pharmacology , Inflammation/metabolism , Inflammation/pathology , Suppressor of Cytokine Signaling 3 Protein/metabolism , Adenylate Kinase/metabolism , Animals , Cell Death/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Culture Media, Conditioned/pharmacology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Enzyme Activation/drug effects , Inflammation Mediators/metabolism , Lipopolysaccharides , Male , Mice , Microglia/drug effects , Microglia/metabolism , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Nitric Oxide Synthase Type II/metabolism , Protein Kinase C-delta/metabolism , Protein Transport/drug effects
18.
Article in English | MEDLINE | ID: mdl-25136372

ABSTRACT

Acute lymphoblastic leukemia (ALL) is a cancer that immature white blood cells continuously overproduce in the bone marrow. These cells crowd out normal cells in the bone marrow bringing damage and death. Methotrexate (MTX) is a drug used in the treatment of various cancer and autoimmune diseases. In particular, for the treatment of childhood acute lymphoblastic leukemia, it had significant effect. MTX competitively inhibits dihydrofolate reductase (DHFR), an enzyme that participates in the tetrahydrofolate synthesis so as to inhibit purine synthesis. In addition, its downstream metabolite methotrexate polyglutamates (MTX-PGs) inhibit the thymidylate synthase (TS). Therefore, MTX can inhibit the synthesis of DNA. However, MTX has cytotoxicity and neurotoxin may cause multiple organ injury and is potentially lethal. Thus, the lower toxicity drugs are necessary to be developed. Recently, diseases treatments with Traditional Chinese Medicine (TCM) as complements are getting more and more attention. In this study, we attempted to discover the compounds with drug-like potential for ALL treatment from the components in TCM. We applied virtual screen and QSAR models based on structure-based and ligand-based studies to identify the potential TCM component compounds. Our results show that the TCM compounds adenosine triphosphate, manninotriose, raffinose, and stachyose could have potential to improve the side effects of MTX for ALL treatment.

19.
Molecules ; 19(7): 8820-39, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24972270

ABSTRACT

Increasing evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play a key role in neurodegeneration. Fisetin, a plant flavonol commonly found in fruits and vegetables, is frequently added to nutritional supplements due to its antioxidant properties. In the present study, treatment with fisetin inhibited microglial cell migration and ROS (reactive oxygen species) production. Treatment with fisetin also effectively inhibited LPS plus IFN-γ-induced nitric oxide (NO) production, and inducible nitric oxide synthase (iNOS) expression in microglial cells. Furthermore, fisetin also reduced expressions of iNOS and NO by stimulation of peptidoglycan, the major component of the Gram-positive bacterium cell wall. Fisetin also inhibited the enhancement of LPS/IFN-γ- or peptidoglycan-induced inflammatory mediator IL (interlukin)-1 ß expression. Besides the antioxidative and anti-inflammatory effects of fisetin, our study also elucidates the manner in fisetin-induced an endogenous anti-oxidative enzyme HO (heme oxygenase)-1 expression. Moreover, the regulatory molecular mechanism of fisetin-induced HO-1 expression operates through the PI-3 kinase/AKT and p38 signaling pathways in microglia. Notably, fisetin also significantly attenuated inflammation-related microglial activation and coordination deficit in mice in vivo. These findings suggest that fisetin may be a candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Flavonoids/pharmacology , Microglia/immunology , Neuroprotective Agents/pharmacology , Animals , Cell Line , Cell Movement/drug effects , Drug Evaluation, Preclinical , Flavonols , Heme Oxygenase-1/metabolism , Lipopolysaccharides/pharmacology , Male , Membrane Proteins/metabolism , Mice, Inbred ICR , Microglia/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Reactive Oxygen Species/metabolism , Up-Regulation
20.
PLoS One ; 9(3): e91167, 2014.
Article in English | MEDLINE | ID: mdl-24621589

ABSTRACT

BACKGROUND/OBJECTIVE: Nicardipine is a calcium channel blocker that has been widely used to control blood pressure in severe hypertension following events such as ischemic stroke, traumatic brain injury, and intracerebral hemorrhage. However, accumulating evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play important roles in neurodegeneration, and the effect of nicardipine on microglial activation remains unresolved. METHODOLOGY/PRINCIPAL FINDINGS: In the present study, using murine BV-2 microglia, we demonstrated that nicardipine significantly inhibits microglia-related neuroinflammatory responses. Treatment with nicardipine inhibited microglial cell migration. Nicardipine also significantly inhibited LPS plus IFN-γ-induced release of nitric oxide (NO), and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Furthermore, nicardipine also inhibited microglial activation by peptidoglycan, the major component of the Gram-positive bacterium cell wall. Notably, nicardipine also showed significant anti-neuroinflammatory effects on microglial activation in mice in vivo. CONCLUSION/SIGNIFICANCE: The present study is the first to report a novel inhibitory role of nicardipine on neuroinflammation and provides a new candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.


Subject(s)
Calcium Channel Blockers/pharmacology , Microglia/drug effects , Neuroprotective Agents/pharmacology , Nicardipine/pharmacology , Animals , Blood Pressure/drug effects , Calcium Channel Blockers/therapeutic use , Cell Line , Cytokines/metabolism , Gene Expression Regulation/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Mice , Microglia/metabolism , Microglia/pathology , Neuroprotective Agents/therapeutic use , Nicardipine/therapeutic use , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...