Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 15(13)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37444518

ABSTRACT

Melanoma is the leading cause of death from cutaneous malignancy. While targeted therapy and immunotherapy with checkpoint inhibitors have significantly decreased the mortality rate of this disease, advanced melanoma remains a therapeutic challenge. Here, we confirmed that interferon-gamma (IFN-γ)-induced PD-L1 expression in melanoma cell lines. This increased expression was down-regulated by the reduction in phosphorylated STAT3 signaling via MET tyrosine kinase inhibitor treatment. Furthermore, immunoprecipitation and confocal immunofluorescence microscopy analysis reveals MET and PD-L1 protein-protein interaction and colocalization on the cell surface membrane of melanoma cells. Together, these findings demonstrate that the IFN-γ-induced PD-L1 expression in melanoma cells is negatively regulated by MET inhibition through the JAK/STAT3 signaling pathway and establish the colocalization and interaction between an RTK and a checkpoint protein in melanoma cells.

2.
Carcinogenesis ; 41(11): 1518-1528, 2020 11 13.
Article in English | MEDLINE | ID: mdl-32602900

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a significant risk factor for lung cancer. One potential mechanism through which COPD contributes to lung cancer development could be through generation of an immunosuppressive microenvironment that allows tumor formation and progression. In this study, we compared the status of immune cells and immune checkpoint proteins in lung tumors induced by the tobacco smoke carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) or NNK + lipopolysaccharide (LPS), a model for COPD-associated lung tumors. Compared with NNK-induced lung tumors, NNK+LPS-induced lung tumors exhibited an immunosuppressive microenvironment characterized by higher relative abundances of PD-1+ tumor-associated macrophages, PD-L1+ tumor cells, PD-1+ CD4 and CD8 T lymphocytes and FOXP3+ CD4 and CD8 T lymphocytes. Also, these markers were more abundant in the tumor tissue than in the surrounding 'normal' lung tissue of NNK+LPS-induced lung tumors. PD-L1 expression in lung tumors was associated with IFNγ/STAT1/STAT3 signaling axis. In cell line models, PD-L1 expression was found to be significantly enhanced in phorbol-12-myristate 13-acetate activated THP-1 human monocytes (macrophages) treated with LPS or incubated in conditioned media (CM) generated by non-small cell lung cancer (NSCLC) cells. Similarly, when NSCLC cells were incubated in CM generated by activated THP-1 cells, PD-L1 expression was upregulated in EGFR- and ERK-dependent manner. Overall, our observations indicate that COPD-like chronic inflammation creates a favorable immunosuppressive microenvironment for tumor development and COPD-associated lung tumors might show a better response to immune checkpoint therapies.


Subject(s)
B7-H1 Antigen/metabolism , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Inflammation/complications , Programmed Cell Death 1 Receptor/metabolism , Pulmonary Disease, Chronic Obstructive/complications , Tumor Microenvironment/immunology , Animals , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinoma, Non-Small-Cell Lung/etiology , Carcinoma, Non-Small-Cell Lung/metabolism , Female , Humans , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred A , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology
3.
Mol Carcinog ; 58(3): 321-333, 2019 03.
Article in English | MEDLINE | ID: mdl-30365189

ABSTRACT

Although members of the hyaluronan (HA)-CD44/HA-mediated motility receptor (RHAMM) signaling pathway have been shown to be overexpressed in lung cancer, their role in lung tumorigenesis is unclear. In the present study, we first determined levels of HA and its receptors CD44 and RHAMM in human non-small cell lung cancer (NSCLC) cells and stromal cells as well as mouse lung tumors. Subsequently, we examined the role of HA-CD44/RHAMM signaling pathway in mediating the proliferation and survival of NSCLC cells and the cross-talk between NSCLC cells and normal human lung fibroblasts (NHLFs)/lung cancer-associated fibroblasts (LCAFs). The highest levels of HA and CD44 were observed in NHLFs/LCAFs followed by NSCLC cells, whereas THP-1 monocytes/macrophages showed negligible levels of both HA and CD44. Simultaneous silencing of HA synthase 2 (HAS2) and HAS3 or CD44 and RHAMM suppressed cell proliferation and survival as well as the EGFR/AKT/ERK signaling pathway. Exogenous HA partially rescued the defect in cell proliferation and survival. Moreover, conditioned media (CM) generated by NHLFs/LCAFs enhanced the proliferation of NSCLC cells in a HA-dependent manner as treatment of NHLFs and LCAFs with HAS2 siRNA, 4-methylumbelliferone, an inhibitor of HASs, LY2228820, an inhibitor of p38MAPK, or treatment of A549 cells with CD44 blocking antibody suppressed the effects of the CM. Upon incubation in CM generated by A549 cells or THP-1 macrophages, NHLFs/LCAFs secreted higher concentrations of HA. Overall, our findings indicate that targeting the HA-CD44/RHAMM signaling pathway could be a promising approach for the prevention and therapy of lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Culture Media, Conditioned/pharmacology , Extracellular Matrix Proteins/metabolism , Fibroblasts/pathology , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice
4.
Proc Natl Acad Sci U S A ; 115(33): E7871-E7880, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061404

ABSTRACT

Smyd1, a muscle-specific histone methyltransferase, has established roles in skeletal and cardiac muscle development, but its role in the adult heart remains poorly understood. Our prior work demonstrated that cardiac-specific deletion of Smyd1 in adult mice (Smyd1-KO) leads to hypertrophy and heart failure. Here we show that down-regulation of mitochondrial energetics is an early event in these Smyd1-KO mice preceding the onset of structural abnormalities. This early impairment of mitochondrial energetics in Smyd1-KO mice is associated with a significant reduction in gene and protein expression of PGC-1α, PPARα, and RXRα, the master regulators of cardiac energetics. The effect of Smyd1 on PGC-1α was recapitulated in primary cultured rat ventricular myocytes, in which acute siRNA-mediated silencing of Smyd1 resulted in a greater than twofold decrease in PGC-1α expression without affecting that of PPARα or RXRα. In addition, enrichment of histone H3 lysine 4 trimethylation (a mark of gene activation) at the PGC-1α locus was markedly reduced in Smyd1-KO mice, and Smyd1-induced transcriptional activation of PGC-1α was confirmed by luciferase reporter assays. Functional confirmation of Smyd1's involvement showed an increase in mitochondrial respiration capacity induced by overexpression of Smyd1, which was abolished by siRNA-mediated PGC-1α knockdown. Conversely, overexpression of PGC-1α rescued transcript expression and mitochondrial respiration caused by silencing Smyd1 in cardiomyocytes. These findings provide functional evidence for a role of Smyd1, or any member of the Smyd family, in regulating cardiac energetics in the adult heart, which is mediated, at least in part, via modulating PGC-1α.


Subject(s)
DNA-Binding Proteins/metabolism , Energy Metabolism/physiology , Histone-Lysine N-Methyltransferase/metabolism , Mitochondria, Heart/metabolism , Muscle Proteins/metabolism , Myocardium/enzymology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Gene Expression Regulation , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Mice , Mice, Knockout , Mitochondria, Heart/genetics , Muscle Proteins/genetics , PPAR alpha/biosynthesis , PPAR alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Retinoid X Receptor alpha/biosynthesis , Retinoid X Receptor alpha/genetics , Transcription Factors/genetics
5.
Diabetes ; 65(9): 2639-51, 2016 09.
Article in English | MEDLINE | ID: mdl-27284109

ABSTRACT

Obesity and insulin resistance are associated with oxidative stress (OS). The causal role of adipose OS in the pathogenesis of these conditions is unknown. To address this issue, we generated mice with an adipocyte-selective deletion of manganese superoxide dismutase (MnSOD). When fed a high-fat diet (HFD), the AdSod2 knockout (KO) mice exhibited less adiposity, reduced adipocyte hypertrophy, and decreased circulating leptin. The resistance to diet-induced adiposity was the result of an increased metabolic rate and energy expenditure. Furthermore, palmitate oxidation was elevated in the white adipose tissue (WAT) and brown adipose tissue of AdSod2 KO mice fed an HFD, and the expression of key fatty acid oxidation genes was increased. To gain mechanistic insight into the increased fat oxidation in HFD-fed AdSod2 KO mice, we quantified the mitochondrial function and mitochondrial content in WAT and found that MnSOD deletion increased mitochondrial oxygen consumption and induced mitochondrial biogenesis. This effect was preserved in cultured adipocytes from AdSod2 KO mice in vitro. As expected from the enhanced fat oxidation, circulating levels of free fatty acids were reduced in the HFD-fed AdSod2 KO mice. Finally, HFD-fed AdSod2 KO mice were protected from hepatic steatosis, adipose tissue inflammation, and glucose and insulin intolerance. Taken together, these results demonstrate that MnSOD deletion in adipocytes triggered an adaptive stress response that activated mitochondrial biogenesis and enhanced mitochondrial fatty acid oxidation, thereby preventing diet-induced obesity and insulin resistance.


Subject(s)
Adipocytes/metabolism , Diet, High-Fat/adverse effects , Mitochondria/metabolism , Obesity/metabolism , Superoxide Dismutase/metabolism , Adiponectin/genetics , Animals , Blotting, Western , Calorimetry, Indirect , Fluorescent Antibody Technique , Mice, Knockout , Obesity/etiology , Oxidative Stress/genetics , Oxidative Stress/physiology , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Palmitates/metabolism , Promoter Regions, Genetic/genetics , Reverse Transcriptase Polymerase Chain Reaction , Superoxide Dismutase/deficiency
6.
J Control Release ; 207: 154-62, 2015 Jun 10.
Article in English | MEDLINE | ID: mdl-25817008

ABSTRACT

The combinatorial peptidergic therapy of islet amyloid polypeptide (IAPP) and leptin (LEP) analogues was once an optimistic option in treating obese animals and patients. However, the need for frequent administrations and its negative side effects prevent it from being a viable choice. Here, we developed a combinatorial gene therapy of IAPP and LEP, where two genes are inserted into a single plasmid with self-cleaving furin and 2A sites to treat diet-induced obese (DIO) mice. The developed plasmid DNA (pDNA) individually produced both IAPP and LEP peptides in vitro and in vivo. The pDNA was delivered with a non-viral polymeric carrier, and its once-a-week administrations demonstrated a synergistic loss of body weight and significant reductions of fat mass, blood glucose, and lipid levels in DIO mice. The results suggest that the combinatorial gene therapy would have higher potential than the peptidergic approach for future translation due to its improved practicability.


Subject(s)
Diet, High-Fat , Gene Transfer Techniques , Genetic Therapy/methods , Leptin/biosynthesis , Obesity/therapy , Polymers/chemistry , Adiposity , Animals , Blood Glucose/metabolism , Disease Models, Animal , Eating , HEK293 Cells , Humans , Islet Amyloid Polypeptide/biosynthesis , Islet Amyloid Polypeptide/genetics , Leptin/genetics , Lipids/blood , Male , Mice, Inbred C57BL , Obesity/blood , Obesity/genetics , Plasmids/genetics , Plasmids/metabolism , Time Factors , Transfection , Weight Loss
7.
Diabetes ; 61(12): 3260-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22912419

ABSTRACT

These studies investigate the role of uncoupling protein 3 (UCP3) in cardiac energy metabolism, cardiac O(2) consumption (MVO(2)), cardiac efficiency (CE), and mitochondrial uncoupling in high fat (HF)-fed or leptin-deficient mice. UCP3KO and wild-type (WT) mice were fed normal chow or HF diets for 10 weeks. Substrate utilization rates, MVO(2), CE, and mitochondrial uncoupling were measured in perfused working hearts and saponin-permeabilized cardiac fibers, respectively. Similar analyses were performed in hearts of ob/ob mice lacking UCP3 (U3OB mice). HF increased cardiac UCP3 protein. However, fatty acid (FA) oxidation rates were similarly increased by HF diet in WT and UCP3KO mice. By contrast, MVO(2) increased in WT, but not in UCP3KO with HF, leading to increased CE in UCP3KO mice. Consistent with increased CE, mitochondrial coupling was increased in the hearts of HF-fed UCP3KO mice. Unexpectedly, UCP3 deletion in ob/ob mice reduced FA oxidation but had no effect on MVO(2) or CE. In addition, FA-induced mitochondrial uncoupling was similarly enhanced in U3OB compared with ob/ob hearts and was associated with elevated mitochondrial thioesterase-1 protein content. These studies show that although UCP3 may mediate mitochondrial uncoupling and reduced CE after HF feeding, it does not mediate uncoupling in leptin-deficient states.


Subject(s)
Energy Metabolism/physiology , Ion Channels/metabolism , Leptin/deficiency , Mitochondrial Proteins/metabolism , Oxygen Consumption/physiology , Animals , Dietary Fats/adverse effects , Energy Metabolism/genetics , Ion Channels/genetics , Leptin/genetics , Male , Mice , Mice, Knockout , Mitochondria , Mitochondrial Proteins/genetics , Oxygen Consumption/genetics , Palmitoyl-CoA Hydrolase/metabolism , Uncoupling Protein 3
8.
Endocrinology ; 153(6): 2677-88, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22510273

ABSTRACT

This study sought to elucidate the relationship between skeletal muscle mitochondrial dysfunction, oxidative stress, and insulin resistance in two mouse models with differential susceptibility to diet-induced obesity. We examined the time course of mitochondrial dysfunction and insulin resistance in obesity-prone C57B and obesity-resistant FVB mouse strains in response to high-fat feeding. After 5 wk, impaired insulin-mediated glucose uptake in skeletal muscle developed in both strains in the absence of any impairment in proximal insulin signaling. Impaired mitochondrial oxidative capacity preceded the development of insulin resistant glucose uptake in C57B mice in concert with increased oxidative stress in skeletal muscle. By contrast, mitochondrial uncoupling in FVB mice, which prevented oxidative stress and increased energy expenditure, did not prevent insulin resistant glucose uptake in skeletal muscle. Preventing oxidative stress in C57B mice treated systemically with an antioxidant normalized skeletal muscle mitochondrial function but failed to normalize glucose tolerance and insulin sensitivity. Furthermore, high fat-fed uncoupling protein 3 knockout mice developed increased oxidative stress that did not worsen glucose tolerance. In the evolution of diet-induced obesity and insulin resistance, initial but divergent strain-dependent mitochondrial adaptations modulate oxidative stress and energy expenditure without influencing the onset of impaired insulin-mediated glucose uptake.


Subject(s)
Energy Metabolism/physiology , Mitochondria, Muscle/physiology , Obesity/physiopathology , Oxidative Stress/physiology , Adaptation, Physiological , Adenosine Triphosphate/metabolism , Animals , Blood Glucose/metabolism , Blotting, Western , Diet, High-Fat/adverse effects , Diglycerides/metabolism , Female , Insulin/blood , Ion Channels/genetics , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Muscle/genetics , Mitochondria, Muscle/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle, Skeletal/metabolism , Obesity/etiology , Obesity/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Time Factors , Triglycerides/metabolism , Uncoupling Protein 3
9.
Oncol Rep ; 27(3): 842-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22024718

ABSTRACT

A stable nitroxide 4-hydroxy-2,2,6,6-tetramethylpiperidine-N-osyl (Tempol) is widely used as an antioxidant in vitro and in vivo. In this study, we investigated the effects of Tempol on the growth of As4.1 juxtaglomerular cells in relation to cell cycle and cell death. Tempol dose-dependently decreased the growth of As4.1 cells with an IC50 of ~1 mM at 48 h. DNA flow cytometry analysis and BrdU staining indicated that Tempol induced S phase arrest, which is accompanied by a downregulation of cyclin A. Tempol also induced apoptotic cell death, which was accompanied by the loss of mitochondrial membrane potential (MMP; ∆Ψm), an activation of caspase-3 and cleavage of poly(ADP-ribose)polymerase-1 (PARP-1). Furthermore, Tempol increased reactive oxygen species (ROS) levels, and the phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK). MEK and JNK inhibitors significantly attenuated a growth inhibition in Tempol-treated As4.1 cells. In conclusion, Tempol inhibited the growth of As4.1 cells via cell cycle arrest and apoptosis. Tempol also activated ERK and JNK signaling, which was responsible for cell growth inhibition. Our present data provide useful information for the toxicological effects of Tempol in juxtaglomerular cells in relation to cell growth inhibition and cell death.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cyclic N-Oxides/pharmacology , Juxtaglomerular Apparatus/drug effects , Animals , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cyclin A/metabolism , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Glutathione/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Phosphorylation/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , S Phase/drug effects , Signal Transduction/drug effects , Spin Labels
10.
Int J Mol Med ; 27(4): 575-81, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21258765

ABSTRACT

Carbonyl cyanide p-(trifluoromethoxy) phenylhydrazone (FCCP) is an uncoupler of mitochondrial oxidative phosphorylation in eukaryotic cells. In the present study, we investigated the involvement of reactive oxygen species (ROS) and glutathione (GSH) in FCCP-induced As4.1 juxtaglomerular cell death. Intracellular ROS levels were decreased by FCCP at the early time points (10-150 min) and increased at 48 h. FCCP inhibited the activity of Mn-superoxide dismutase (Mn-SOD) via down-regulating its protein expression. Ebselen (an antioxidant) significantly attenuated ROS levels in FCCP-treated cells, but did not prevent FCCP-induced cell death. Moreover, intracellular GSH content was rapidly diminished within 10 min of FCCP treatment, which was accompanied by a reduction of the mitochondrial membrane potential [MMP (∆ψm)]. L-buthionine sulfoximine (BSO, a GSH synthesis inhibitor) significantly augmented As4.1 cell death by FCCP. However, N-acetylcysteine (NAC, a GSH precursor and antioxidant) attenuated GSH depletion, MMP (∆ψm) loss and cell death in FCCP-treated As4.1 cells. In addition, NAC increased Mn-SOD activity and decreased ROS levels in FCCP-treated As4.1 cells. In conclusion, these results suggest that compared to ROS levels, intracellular GSH levels are more closely linked to FCCP-induced apoptosis in As4.1 juxtaglomerular cells.


Subject(s)
Apoptosis/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Glutathione/metabolism , Intracellular Space/metabolism , Juxtaglomerular Apparatus/drug effects , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Animals , Buthionine Sulfoximine/metabolism , Buthionine Sulfoximine/pharmacology , Catalase/metabolism , Cell Death/drug effects , Cell Line , Juxtaglomerular Apparatus/metabolism , Mice , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
11.
Anticancer Res ; 30(7): 2863-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20683024

ABSTRACT

Carbonyl cyanide p-(trifluoromethoxy) phenylhydrazone (FCCP) is an uncoupler of mitochondrial oxidative phosphorylation in mitochondria. This study evaluated the effects of FCCP on the growth of juxtaglomerular As4.1 cells in relation to the cell cycle and apoptosis. FCCP inhibited the growth of As4.1 cells with an IC(50) of approximately 10 muM at 48 hours. DNA flow cytometry indicated that FCCP did not induce the specific phase arrests of the cell cycle. This agent efficiently reduced mitochondrial membrane potential (MMP; DeltaPsi(m)) levels within 1 hour and dose-dependently induced loss of MMP (DeltaPsi(m)) at 48 h. FCCP also induced apoptosis in As4.1 cells, as evidenced by sub-G(1) cells and annexin V binding assay. This apoptotic process was accompanied by caspase-3 activation and PARP cleavage. Although caspase-3 inhibitor significantly reduced the activity of caspase-3, all of the caspase inhibitors tested in this study failed to rescue As4.1 cells from FCCP-induced cell death. In conclusion, this study demonstrated that FCCP, as a mitochondria-damaging agent, potently induces apoptosis in As4.1 juxtaglomerular cells via a caspase-independent manner.


Subject(s)
Apoptosis/drug effects , Carbonyl Cyanide p-Trifluoromethoxyphenylhydrazone/pharmacology , Juxtaglomerular Apparatus/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis/physiology , Caspase 3/metabolism , Caspase Inhibitors , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Juxtaglomerular Apparatus/cytology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/physiology , Uncoupling Agents/pharmacology
12.
Anticancer Res ; 30(6): 2107-12, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20651358

ABSTRACT

MG132 as a proteasome inhibitor can induce apoptotic cell death through formation of reactive oxygen species (ROS). In this study, the effects of N-acetyl cysteine (NAC; an antioxidant) on MG132-induced HeLa cell death in relation to ROS and glutathione (GSH) were investigated. MG132 induced cell growth inhibition and apoptosis in HeLa cells, which was accompanied by the loss of mitochondrial membrane potential (MMP; Delta Psi(m)), activation of caspase-3 and poly (ADP-ribose) polymerase (PARP) cleavage. MG132 increased ROS levels, including O(2)(*-), and GSH depleted cell numbers of HeLa cells. NAC reduced the number of annexin V-positive cells and MMP (Delta Psi(m)) loss by MG132. In addition, NAC significantly reduced the ROS level and prevented GSH depletion. In conclusion, NAC prevented MG132-induced HeLa cell death via decreasing ROS and preventing GSH depletion.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Glutathione/metabolism , Leupeptins/pharmacology , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Caspase 3/physiology , Cell Cycle/drug effects , Cell Proliferation/drug effects , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Proteasome Inhibitors
13.
Drug Chem Toxicol ; 33(4): 367-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20545600

ABSTRACT

MG132, as a proteasome inhibitor, has been shown to induce apoptotic cell death through the formation of reactive oxygen species (ROS). In this study, we investigated the effects of MG132 and/or MAPK inhibitors on As4.1 juxtaglomerular cells in relation to cell growth, cell death, ROS, and glutathione (GSH) levels. MG132 inhibited the growth of As4.1 cells and induced cell death, accompanied by the loss of mitochondrial membrane potential (MMP; DeltaPsi(m)) and activation of caspase-3 and -8. MG132 increased ROS levels, and GSH depleted cell numbers. The MEK inhibitor slightly reduced cell growth and caspase-3 activity in MG132-treated As4.1 cells and mildly increased MMP (DeltaPsi(m)) loss and O(2)(*-) level. However, it did not increase apoptosis and GSH depletion. The JNK inhibitor did not strongly influence cell growth, cell death, and GSH depletion by MG132, but increased caspase-3 activity, MMP (DeltaPsi(m)) loss, and O(2)(*-) level. Treatment with the p38 inhibitor magnified cell-growth inhibition and apoptosis by MG132. This agent also strongly increased caspase-8 activity, MMP (DeltaPsi(m)) loss, O(2)(*-) level, and GSH depletion. Conclusively, the p38 inhibitor strongly intensified cell death in MG132-treated As4.1 cells. The changes of GSH content by MG132 and/or MAPK inhibitors were closely related to the death of As4.1 cells.


Subject(s)
Apoptosis/drug effects , Glutathione/metabolism , Juxtaglomerular Apparatus/drug effects , Leupeptins/pharmacology , Proteasome Inhibitors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Caspase 3/metabolism , Caspase 8/metabolism , Cell Death/drug effects , Cell Line, Tumor , Juxtaglomerular Apparatus/cytology , Juxtaglomerular Apparatus/enzymology , Juxtaglomerular Apparatus/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice , Reactive Oxygen Species/metabolism
14.
Arch Toxicol ; 84(9): 689-98, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20449726

ABSTRACT

The proteasome inhibitor MG132 has been shown to induce apoptotic cell death through the formation of reactive oxygen species (ROS). Here, we investigated the molecular mechanisms of MG132 in As4.1 juxtaglomerular cell death in relation to apoptosis and levels of ROS and glutathione (GSH). MG132 inhibited the growth of As4.1 cells with an IC(50) of approximately 0.3-0.4 microM at 48 h and induced cell death, accompanied by the loss of mitochondrial membrane potential (MMP; Psi(m)), Bcl-2 decrease, activations of caspase-3 and caspase-8, and PARP cleavage. MG132 increased intracellular ROS levels and GSH-depleted cell numbers. However, caspase inhibitors, especially Z-VAD (pan-caspase inhibitor) intensified cell growth inhibition, cell death, MMP (Psi(m)) loss, and Bcl-2 decrease in MG132-treated As4.1 cells. Z-VAD also slightly intensified increases in ROS levels and GSH depletion in MG132-treated As4.1 cells. In conclusion, MG132 reduced the growth of As4.1 cells via caspase-independent apoptosis. The changes in ROS and GSH levels by MG132 and caspase inhibitors partially influenced the growth inhibition and death of As4.1 cells.


Subject(s)
Apoptosis , Juxtaglomerular Apparatus/metabolism , Leupeptins/metabolism , Proteasome Inhibitors , Animals , Caspase Inhibitors , Cell Death , Cell Line , Cysteine Proteinase Inhibitors/metabolism , Glutathione/metabolism , Juxtaglomerular Apparatus/cytology , Membrane Potential, Mitochondrial , Mice , Proteasome Endopeptidase Complex/metabolism , Reactive Oxygen Species/metabolism
15.
Cancer Genet Cytogenet ; 199(2): 81-8, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20471510

ABSTRACT

MG132 (carbobenzoxy-Leu-Leu-leucinal) as a proteasome inhibitor has been shown to induce apoptotic cell death through formation of reactive oxygen species (ROS). In this study, we investigated the effects of MEK (mitogen-activated protein [MAP] kinase or extracellular signal-regulated kinase [ERK] kinase) or p38 inhibitor on MG132-treated Calu-6 lung cancer cells in relation to cell growth, cell death, ROS, and glutathione (GSH) levels. Treatment with 10 mumol/L MG132 inhibited the growth of Calu-6 cells at 24 hours. MG132 induced apoptosis in Calu-6 cells, which was accompanied by the loss of mitochondrial membrane potential (MMP; DeltaPsi(m)). ROS were increased in MG132-treated Calu-6 cells. MG132 also induced GSH depletion in Calu-6 cells. Treatment with MEK inhibitor did not significantly affect cell growth, cell death, ROS, and GSH levels in MG132-treated Calu-6 cells. Furthermore, MG132 increased the phosphorylation of p38 in Calu-6 cells at 1 and 24 hours. Treatment with p38 inhibitor significantly prevented cell growth inhibition, MMP (DeltaPsi(m)) loss and apoptosis in MG132-treated Calu-6 cells. This inhibitor increased ROS level and decreased GSH depletion in these cells. In conclusion, p38 inhibitor partially prevented Calu-6 cell death by MG132, which might be affected by GSH level changes.


Subject(s)
Adenocarcinoma/drug therapy , Cysteine Proteinase Inhibitors/toxicity , Imidazoles/therapeutic use , Leupeptins/toxicity , Lung Neoplasms/drug therapy , Pyridines/therapeutic use , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis/drug effects , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Flavonoids/pharmacology , Glutathione/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
16.
Anticancer Res ; 30(3): 879-85, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20393010

ABSTRACT

MG132, a proteasome inhibitor, has been shown to induce apoptotic cell death through formation of reactive oxygen species (ROS). Here, we evaluated the effects of MG132 on the growth of endothelial cells, especially calf pulmonary artery endothelial cells (CPAECs). MG132 dose-dependently inhibited the growth of CPAECSs and human umbilical vein endothelial cells (HUVECs) at 24 hours. MG132 also induced apoptosis in both cell lines, which was accompanied by the loss of mitochondrial membrane potential. All the tested caspase inhibitors (pan-caspase, caspase-3, -8 and -9 inhibitor) significantly rescued CPAECs from MG132-induced cell death. MG132 increased ROS level and GSH depleted cell numbers of CPAECs. None of the caspase inhibitors reduced ROS level in MG132-treated CPAECs but did reduce apoptosis in these cells. In conclusion, MG132 inhibited the growth of endothelial cells, especially CPAECs via caspase-dependent apoptosis. MG132-induced CPAEC death was related to GSH depletion rather than a change in ROS level.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Endothelial Cells/drug effects , Leupeptins/pharmacology , Animals , Apoptosis/physiology , Caspase Inhibitors , Cattle , Cell Growth Processes/drug effects , Cell Line , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/metabolism , Glutathione/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Pulmonary Artery/cytology , Reactive Oxygen Species/metabolism , Umbilical Veins/cytology
17.
Int J Mol Med ; 25(6): 937-44, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428799

ABSTRACT

Propyl gallate (PG) as a synthetic antioxidant exerting a variety of effects on tissue and cell functions. We evaluated the effects of PG on the growth of endothelial cells, especially calf pulmonary artery endothelial cells (CPAEC) in relation to apoptosis. PG dose-dependently inhibited the growth of CPAEC and human umbilical vein endothelial cells (HUVEC) at 24 h. The susceptibility of CPAEC to PG was higher than that of HUVEC. PG induced apoptosis in CPAEC, which was accompanied by the loss of mitochondrial membrane potential (MMP; DeltaPsim). The tested caspase inhibitors (pan-caspase, caspase-3, -8 or -9 inhibitor) did not rescue CPAEC from PG-induced cell death but instead slightly enhanced the cell death. PG increased reactive oxygen species (ROS) level in CPAEC. The caspase inhibitors did not significantly change the ROS level. Furthermore, PG increased the GSH depleted cell number and decreased GSH level in CPAEC. The tested caspase inhibitors did not significantly change the number in PG-treated CPAEC. Each caspase inhibitor differently alters GSH levels in CPAEC. In conclusion, PG inhibited the growth of endothelial cells, especially CPAEC via caspase-independent apoptosis. PG-induced CPAEC death was accompanied by ROS increase and GSH depletion.


Subject(s)
Apoptosis/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Propyl Gallate/pharmacology , Pulmonary Artery/cytology , Animals , Caspase Inhibitors , Caspases/metabolism , Cattle , Cell Proliferation/drug effects , Endothelial Cells/enzymology , Glutathione/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Propyl Gallate/chemistry , Reactive Oxygen Species/metabolism , Umbilical Veins/cytology
18.
Oncol Res ; 18(7): 315-22, 2010.
Article in English | MEDLINE | ID: mdl-20377132

ABSTRACT

MG132, as a proteasome inhibitor, can induce apoptotic cell death through formation of reactive oxygen species (ROS). In this study, we investigated the effects of MAPK (MEK, JNK, and p38) inhibitors on MG132-treated A549 lung cancer cells in relation to cell growth, cell death, ROS, and glutathione (GSH) levels. Treatment with 10 microM MG132 inhibited the growth of A549 cells at 24 h. MG132 also induced apoptosis, which was accompanied by the loss of mitochondrial membrane potential (MMP; deltapsi(m)). ROS were not increased in MG132-treated A549 cells. MG132 increased GSH-depleted cell numbers and decreased GSH levels. MEK and JNK inhibitors did not strongly affect cell growth, cell death, ROS, and GSH levels in MG132-treated A549 cells. In contrast, p38 inhibitor reduced cell growth inhibition, apoptosis, and MMP (deltapsi(m)) loss by MG132. However, p38 inhibitor did not change ROS level and GSH content. In conclusion, MG132 inhibited the growth of A549 cells via apoptosis without formation of ROS. Treatment with p38 inhibitor rescued some cells from MG132-induced apotposis, which was not affected by ROS and GSH level changes.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Leupeptins/pharmacology , Lung Neoplasms/pathology , Proteasome Inhibitors , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Glutathione/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Food Chem Toxicol ; 48(5): 1334-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20197077

ABSTRACT

Gallic acid (GA) is widely distributed in various plants and foods, and its various biological effects have been reported. Here, we evaluated the effects of GA on HeLa cells in relation to cell growth inhibition and death. HeLa cell growth was diminished with an IC(50) of approximately 80 microM GA at 24h whereas an IC(50) of GA in human umbilical vein endothelial cells (HUVEC) was approximately 400 microM. GA-induced apoptosis and/or necrosis in HeLa cells and HUVEC, which was accompanied by the loss of mitochondrial membrane potential (MMP; DeltaPsi(m)). The percentages of MMP (DeltaPsi(m)) loss cells and death cells were lower in HUVEC than HeLa cells. All the tested caspase inhibitors (pan-caspase, caspase-3, -8 or -9 inhibitor) significantly rescued HeLa cells from GA-induced cell death. GA increased reactive oxygen species (ROS) level and GSH (glutathione) depleted cell number in HeLa cells. Caspase inhibitors reduced GSH depleted cell number but not ROS level in GA-treated HeLa cells. In conclusion, GA inhibited the growth of HeLa cells and HUVEC via apoptosis and/or necrosis. The susceptibility of HeLa cells to GA was higher than that of HUVEC. GA-induced HeLa cell death was accompanied by ROS increase and GSH depletion.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Gallic Acid/pharmacology , Necrosis/chemically induced , Uterine Cervical Neoplasms/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Glutathione/metabolism , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Superoxides/metabolism , Umbilical Veins/cytology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology
20.
Oncol Rep ; 23(4): 1153-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20204304

ABSTRACT

Propyl gallate (PG) as a synthetic antioxidant exerts a variety of effects on tissue and cell functions. Here, we evaluated the effects of PG on the growth of HeLa cells in relation to apoptosis and the cell cycle. PG dose-dependently inhibited the growth of HeLa cells with an IC50 of approximately 800 microM at 24 h. DNA flow cytometric analysis indicated that PG significantly induced a G1 phase arrest of the cell cycle along with an increase in the cyclin-dependent kinase inhibitor (CDKI) p27. In addition, PG induced apoptosis, which was accompanied by the loss of mitochondrial membrane potential (MMP; DeltaPsim), activation of caspase-3 and caspase-8 and PARP cleavage. All the tested caspase inhibitors (pan-caspase, caspase-3, -8 or -9 inhibitor) significantly rescued HeLa cells from PG-induced cell death. However, none of the caspase inhibitors prevented the loss of MMP (DeltaPsim) induced by PG. In conclusion, PG inhibited the growth of HeLa cells via caspase-dependent apoptosis as well as a G1 phase arrest of the cell cycle.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , G1 Phase/drug effects , Propyl Gallate/pharmacology , Blotting, Western , Caspases/drug effects , Cell Proliferation/drug effects , Cell Separation , Flow Cytometry , HeLa Cells , Humans , Inhibitory Concentration 50 , Membrane Potential, Mitochondrial/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...