Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
FASEB J ; 36(1): e22096, 2022 01.
Article in English | MEDLINE | ID: mdl-34907600

ABSTRACT

Tuberculosis is a communicable disease caused by Mycobacterium tuberculosis which primarily infects macrophages and establishes intracellular parasitism. A mycobacterial virulence factor Zn2+ metalloprotease 1 (Zmp1) is known to suppress interleukin (IL)-1ß production by inhibiting caspase-1 resulting in phagosome maturation arrest. However, the molecular mechanism of caspase-1 inhibition by Zmp1 is still elusive. Here, we identified GRIM-19 (also known as NDUFA13), an essential subunit of mitochondrial respiratory chain complex I, as a novel Zmp1-binding protein. Using the CRISPR/Cas9 system, we generated GRIM-19 knockout murine macrophage cell line J774.1 and found that GRIM-19 is essential for IL-1ß production during mycobacterial infection as well as in response to NLRP3 inflammasome-activating stimuli such as extracellular ATP or nigericin. We also found that GRIM-19 is required for the generation of mitochondrial reactive oxygen species and NLRP3-dependent activation of caspase-1. Loss of GRIM-19 or forced expression of Zmp1 resulted in a decrease in mitochondrial membrane potential. Our study revealed a previously unrecognized role of GRIM-19 as an essential regulator of NLRP3 inflammasome and a molecular mechanism underlying Zmp1-mediated suppression of IL-1ß production during mycobacterial infection.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Mycobacterium tuberculosis/metabolism , NADH, NADPH Oxidoreductases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Bacterial Proteins , Gene Knockdown Techniques , HEK293 Cells , Humans , Inflammasomes/genetics , Metalloproteases , Mice , Mitochondrial Membranes/metabolism , Mycobacterium tuberculosis/genetics , NADH, NADPH Oxidoreductases/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
2.
Cancer Treat Res Commun ; 28: 100386, 2021.
Article in English | MEDLINE | ID: mdl-34010730

ABSTRACT

Resveratrol (RES), a dietary phenolic compound, was reported to have cancer chemoprotective and chemotherapeutic effects. Earlier we unexpectedly observed that RES has a growth-enhancing effect in some breast cancer cells and can diminish the susceptibility of MDA-MB-231 and SKBR-3 cells to paclitaxel-induced cell death, but this phenomenon is not observed in MCF-7 cells. The present study seeks to determine the mechanism underlying RES's attenuation of paclitaxel cytotoxicity in cancer cells. It is found that RES reduces the anticancer action of paclitaxel only in the human breast cancer cells that express HER3 protein. Treatment of SKBR-3 cells with RES increases HER3 expression in a dose-dependent manner. The induction of HER3 expression by RES confers resistance of breast cancer cells against paclitaxel cytotoxicity. Furthermore, it is observed that the SIRT1-FOXO1 signaling pathway plays an important role in mediating RES-induced upregulation of HER3 expression. In conclusion, the present study reveals the mechanism for RES-induced resistance against paclitaxel in some human breast cancer cells, and it is suggested that the combined use of RES and paclitaxel is not suitable for treating human breast cancer that expresses HER3 protein.


Subject(s)
Antineoplastic Agents, Phytogenic/adverse effects , Breast Neoplasms/drug therapy , Paclitaxel/adverse effects , Protective Agents/pharmacology , Resveratrol/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Forkhead Box Protein O1/genetics , Humans , Receptor, ErbB-3/genetics , Receptor, ErbB-3/metabolism , Signal Transduction/drug effects , Sirtuin 1/genetics
3.
Biochem Biophys Res Commun ; 477(3): 495-502, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27317486

ABSTRACT

Glutathione depletion is a distinct cause underlying many forms of pathogenesis associated with oxidative stress and cytotoxicity. Earlier studies showed that glutamate-induced glutathione depletion in immortalized murine HT22 hippocampal neuronal cells leads to accumulation of reactive oxygen species (ROS) and ultimately cell death, but the precise mechanism underlying these processes is not clear. Here we show that during the induction of glutathione depletion, nitric oxide (NO) accumulation precedes ROS accumulation. While neuronal NO synthase (nNOS) in untreated HT22 cells exists mostly as a monomer, glutathione depletion results in increased formation of the dimer nNOS, accompanied by increases in the catalytic activity. We identified that nNOS dimerization is catalyzed by protein disulfide isomerase (PDI). Inhibition of PDI's isomerase activity effectively abrogates glutathione depletion-induced conversion of monomer nNOS into dimer nNOS, accumulation of NO and ROS, and cytotoxicity. Furthermore, we found that PDI is present in untreated cells in an inactive S-nitrosylated form, which becomes activated following glutathione depletion via S-denitrosylation. These results reveal a novel role for PDI in mediating glutathione depletion-induced oxidative cytotoxicity, as well as its role as a valuable therapeutic target for protection against oxidative cytotoxicity.


Subject(s)
Glutathione/metabolism , Protein Disulfide-Isomerases/metabolism , Animals , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
4.
Microbiol Immunol ; 59(12): 735-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26577130

ABSTRACT

Although the BCG vaccine can prevent tuberculosis (TB) in infants, its ability to prevent adult pulmonary TB is reportedly limited. Therefore, development of a novel effective vaccine against pulmonary TB has become an international research priority. We have previously reported that intranasal vaccination of mice with a mycobacterial heparin-binding hemagglutinin adhesin (HBHA) plus mucosal adjuvant cholera toxin (CT) enhances production of IFN-γ and anti-HBHA antibody and suppresses extrapulmonary bacterial dissemination after intranasal infection with BCG. In the present study, the effects of intranasal HBHA + CT vaccine on murine pulmonary Mycobacterium tuberculosis (Mtb) infection were examined. Intranasal HBHA + CT vaccination alone failed to reduce the bacterial burden in the infected lung. However, a combination vaccine consisting of s.c. BCG priming and an intranasal HBHA + CT booster significantly enhanced protective immunity against pulmonary Mtb infection on day 14 compared with BCG vaccine alone. Further, it was found that intranasal HBHA + CT vaccine enhanced not only IFN-γ but also IL-17A production by HBHA-specific T cells in the lung after pulmonary Mtb infection. Therefore, this combination vaccine may be a good candidate for a new vaccine strategy against pulmonary TB.


Subject(s)
Adhesins, Bacterial/immunology , BCG Vaccine/pharmacology , Lectins/immunology , Lung/immunology , Mycobacterium tuberculosis/immunology , Th17 Cells/immunology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/pharmacology , Animals , Antigens, Bacterial/immunology , BCG Vaccine/immunology , Cholera Toxin/immunology , Female , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Interleukin-17/immunology , Lectins/genetics , Lung/pathology , Mice , Mice, Inbred C57BL , T-Lymphocytes/metabolism , Th1 Cells/immunology , Tuberculosis, Pulmonary/immunology
5.
Sci Rep ; 5: 16712, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26567927

ABSTRACT

Altered sensory experience in early life often leads to remarkable adaptations so that humans and animals can make the best use of the available information in a particular environment. By restricting visual input to a limited range of orientations in young animals, this investigation shows that stimulus selectivity, e.g., the sharpness of tuning of single neurons in the primary visual cortex, is modified to match a particular environment. Specifically, neurons tuned to an experienced orientation in orientation-restricted animals show sharper orientation tuning than neurons in normal animals, whereas the opposite was true for neurons tuned to non-experienced orientations. This sharpened tuning appears to be due to elongated receptive fields. Our results demonstrate that restricted sensory experiences can sculpt the supranormal functions of single neurons tailored for a particular environment. The above findings, in addition to the minimal population response to orientations close to the experienced one, agree with the predictions of a sparse coding hypothesis in which information is represented efficiently by a small number of activated neurons. This suggests that early brain areas adopt an efficient strategy for coding information even when animals are raised in a severely limited visual environment where sensory inputs have an unnatural statistical structure.


Subject(s)
Orientation/physiology , Visual Cortex/physiology , Visual Pathways/physiology , Animals , Cats , Electrophysiological Phenomena , Eye Protective Devices , Optical Imaging , Photic Stimulation
6.
Planta Med ; 81(10): 838-46, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26132849

ABSTRACT

The present study investigates the anticancer effect of ascorbate in MIA-PaCa-2 human pancreatic cancer cells using both in vitro and in vivo models, with a focus on assessing the role of oxidative stress and autophagy as important mechanistic elements in its anticancer actions. We showed that ascorbate suppresses the growth of human pancreatic cancer cells via the induction of oxidative stress and caspase-independent cell death. Ascorbate induces the formation of autophagosomes and the presence of autophagy inhibitors suppresses ascorbate-induced cell death. These data suggest that the induction of autophagosome formation contributes to ascorbate-induced pancreatic cancer cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Ascorbic Acid/pharmacology , Autophagy/drug effects , Membrane Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Beclin-1 , Cell Death/drug effects , Cell Line, Tumor/drug effects , Female , Humans , Membrane Proteins/genetics , Mice, Nude , Microtubule-Associated Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
7.
J Cell Biochem ; 114(1): 192-203, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22903547

ABSTRACT

In a recent study, we showed that eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), two common omega-3 fatty acids, can cause ROS accumulation and subsequently induce caspase-8-dependent apoptosis in human breast cancer cells (Kang et al. [2010], PLoS ONE 5: e10296). In this study, we showed that the pancreas has a unique ability to accumulate EPA at a level markedly higher than several other tissues analyzed. Based on this finding, we sought to further investigate the anticancer actions of EPA and its analog DHA in human pancreatic cancer cells using both in vitro and in vivo models. EPA and DHA were found to induce ROS accumulation and caspase-8-dependent cell death in human pancreatic cancer cells (MIA-PaCa-2 and Capan-2) in vitro. Feeding animals with a diet supplemented with 5% fish oil, which contains high levels of EPA and DHA, also strongly suppresses the growth of MIA-PaCa-2 human pancreatic cancer xenografts in athymic nude mice, by inducing oxidative stress and cell death. In addition, we showed that EPA can concomitantly induce autophagy in these cancer cells, and the induction of autophagy diminishes its ability to induce apoptotic cell death. It is therefore suggested that combination of EPA with an autophagy inhibitor may be a useful strategy in increasing the therapeutic effectiveness in pancreatic cancer.


Subject(s)
Autophagy/drug effects , Caspase 8/metabolism , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/therapeutic use , Pancreatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Caspase 8/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Enzyme Activation/drug effects , Female , Gene Expression/drug effects , Humans , Mice , Mice, Nude , Oxidative Stress/drug effects , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
8.
Brain Res ; 1497: 1-14, 2013 Feb 25.
Article in English | MEDLINE | ID: mdl-23206800

ABSTRACT

Levodopa (L-DOPA) is widely used for symptomatic management in Parkinson's disease. We recently showed that (-)-epigallocatechin-3-gallate, a tea polyphenol, not only inhibits L-DOPA methylation, but also protects against oxidative hippocampal neurodegeneration. In the present study, we sought to determine several other common dietary phenolics, namely, tea catechins [(+)-catechin and (-)-epicatechin] and a representative flavonoid (quercetin), for their ability to modulate L-DOPA methylation and to protect against oxidative hippocampal injury. A combination of in vitro biochemical assays, cell culture-based mechanistic analyses, and in vivo animal models was used. While both tea catechins and quercetin strongly inhibit human liver catechol-O-methyltransferase (COMT)-mediated O-methylation of L-DOPA in vitro, only (+)-catechin exerts a significant inhibition of L-DOPA methylation in both peripheral compartment and striatum in rats. The stronger in vivo effect of (+)-catechin on L-DOPA methylation compared to the other dietary compounds is due to its better bioavailability in vivo. In addition, (+)-catechin strongly reduces glutamate-induced oxidative cytotoxicity in HT22 mouse hippocampal neurons in vitro through inactivation of the nuclear factor-κB signaling pathway. Administration of (+)-catechin also exerts a strong neuroprotective effect in the kainic acid-induced oxidative hippocampal neurodegeneration model in rats. In conclusion, (+)-catechin is a dietary polyphenolic that may have beneficial effects in L-DOPA-based treatment of Parkinson patients by inhibiting L-DOPA methylation plus reducing oxidative neurodegeneration.


Subject(s)
Catechin/pharmacology , Catechol O-Methyltransferase/metabolism , Hippocampus/pathology , Hydroxybenzoates/pharmacology , Nerve Degeneration/prevention & control , Adrenergic Uptake Inhibitors/pharmacology , Analysis of Variance , Animals , Antiparkinson Agents/adverse effects , Antiparkinson Agents/blood , Carbidopa/pharmacology , Chromatography, High Pressure Liquid/methods , Dihydroxyphenylalanine/analogs & derivatives , Dihydroxyphenylalanine/blood , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Fluoresceins , Glial Fibrillary Acidic Protein/metabolism , In Vitro Techniques , Kainic Acid/toxicity , Levodopa/adverse effects , Levodopa/blood , Male , Methylation/drug effects , Mice , Nerve Degeneration/chemically induced , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Reserpine/pharmacology , Time Factors , Tyrosine/analogs & derivatives
9.
Biochem J ; 447(1): 115-23, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22747530

ABSTRACT

E(2) (17ß-oestradiol), a female sex hormone, has important biological functions in a woman's body. The pancreas, often considered a non-classical E(2)-targeting organ, is known to be functionally regulated by E(2), but little is known about how oestrogen actions are regulated in this organ. In the present study we report that PDIp (pancreas-specific protein disulfide isomerase), a protein-folding catalyst, can act as a major intracellular E(2) storage protein in a rat model to modulate the pancreatic tissue level, metabolism and action of E(2). The purified endogenous PDIp from both rat and human pancreatic tissues can bind E(2) with a K(d) value of approximately 150 nM. The endogenous PDIp-bound E(2) accounts for over 80% of the total protein-bound E(2) present in rat and human pancreatic tissues, and this binding protects E(2) from metabolic disposition and prolongs its duration of action. Importantly, we showed in ovariectomized female rats that the E(2) level in the pancreas reaches its highest level (9-fold increase over its basal level) at 24-48 h after a single injection of E(2), and even at 96 h its level is still approximately 5-fold higher. In contrast, the E(2) level in the uterus quickly returns to its basal level at 48 h after reaching its maximal level (approximately 2-fold increase) at 24 h. Taken together, these results show for the first time that PDIp is a predominant intracellular oestrogen storage protein in the pancreas, which offers novel mechanistic insights into the accumulation and action of oestrogen inside pancreatic cells.


Subject(s)
Estrogens/metabolism , Pancreas/metabolism , Protein Disulfide-Isomerases/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Estradiol/metabolism , Female , Humans , Kinetics , Rats , Rats, Sprague-Dawley
10.
Toxicol Appl Pharmacol ; 262(2): 156-66, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22575170

ABSTRACT

Studies in recent years have revealed that excess mitochondrial superoxide production is an important etiological factor in neurodegenerative diseases, resulting from oxidative modifications of cellular lipids, proteins, and nucleic acids. Hence, it is important to understand the mechanism by which mitochondrial oxidative stress causes neuronal death. In this study, the immortalized mouse hippocampal neuronal cells (HT22) in culture were used as a model and they were exposed to menadione (also known as vitamin K(3)) to increase intracellular superoxide production. We found that menadione causes preferential accumulation of superoxide in the mitochondria of these cells, along with the rapid development of mitochondrial dysfunction and cellular ATP depletion. Neuronal death induced by menadione is independent of the activation of the MAPK signaling pathways and caspases. The lack of caspase activation is due to the rapid depletion of cellular ATP. It was observed that two ATP-independent mitochondrial nucleases, namely, AIF and Endo G, are released following menadione exposure. Silencing of their expression using specific siRNAs results in transient suppression (for ~12h) of mitochondrial superoxide-induced neuronal death. While suppression of the mitochondrial superoxide dismutase expression markedly sensitizes neuronal cells to mitochondrial superoxide-induced cytotoxicity, its over-expression confers strong protection. Collectively, these findings showed that many of the observed features associated with mitochondrial superoxide-induced cell death, including caspase independency, rapid depletion of ATP level, mitochondrial release of AIF and Endo G, and mitochondrial swelling, are distinctly different from those of apoptosis; instead they resemble some of the known features of necroptosis.


Subject(s)
Hippocampus/metabolism , Mitochondria/metabolism , Neurodegenerative Diseases/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Superoxides/metabolism , Vitamin K 3/pharmacology , Animals , Apoptosis Inducing Factor/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Line , Endodeoxyribonucleases/metabolism , Hippocampus/drug effects , Hippocampus/pathology , Mice , Microscopy, Electron, Transmission , Mitochondria/drug effects , Mitochondria/ultrastructure , Neurodegenerative Diseases/pathology , Neurons/drug effects , Neurons/pathology , Oxidative Stress/drug effects , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
11.
J Immunotoxicol ; 8(4): 346-58, 2011.
Article in English | MEDLINE | ID: mdl-22017689

ABSTRACT

Estriol (E(3)), an endogenous estrogen predominantly produced during human pregnancy, has been suggested to play an important role in modulating the immune system function during pregnancy. The present study sought to investigate the ability of E(3) to alter splenocyte functions in non-immunized naïve BALB/c female mice and also in mice injected with complete Freund's adjuvant (CFA), and the effect of E(3) was compared with that of 17ß-estradiol (E(2)). When mice were injected with CFA, their spleen weight index (i.e., wet organ wet/whole body weight) was increased by ~ 300%, but co-administration of E(3) almost completely suppressed splenomegaly. E(3) also reduced cytokine production and reduced ERK and p38 activation in both splenocytes and peritoneal exudate cells from CFA-treated animals. In comparison, while E(2) had a similar but slightly weaker effect than E(3) in reducing splenomegaly, it had a rather different effect from E(3) on cytokine production and ERK activation in splenocytes and peritoneal exudate cells from CFA-treated mice. Under naïve immunological conditions, E(3) and E(2) had very similar effects on splenocyte functions. Both of them transiently increased the percentages of splenic CD4(+) and CD8(+) cells. They also increased the proliferation of splenocytes ex vivo, and stimulated production of interferon-γ and interleukin-2. Altogether, these data show that E(3) and E(2) have different effects on splenocyte functions when the animals are under experimentally induced inflammatory conditions.


Subject(s)
Estradiol/pharmacology , Estriol/pharmacology , Estrogens/pharmacology , Freund's Adjuvant/pharmacology , Spleen/drug effects , Animals , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cell Proliferation/drug effects , Cytokines/metabolism , Drug Antagonism , Female , Interferon-gamma/metabolism , Interleukin-2/metabolism , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Spleen/metabolism , Spleen/pathology , Splenomegaly/chemically induced
12.
PLoS One ; 6(8): e24312, 2011.
Article in English | MEDLINE | ID: mdl-21918689

ABSTRACT

BACKGROUND: During a normal cell cycle, the transition from G2 phase to mitotic phase is triggered by the activation of the cyclin B1-dependent Cdc2 kinase. Here we report our finding that treatment of MCF-7 human breast cancer cells with nocodazole, a prototypic microtubule inhibitor, results in strong up-regulation of cyclin B1 and Cdc2 levels, and their increases are required for the development of mitotic prometaphase arrest and characteristic phenotypes. METHODOLOGY/PRINCIPAL FINDINGS: It was observed that there was a time-dependent early increase in cyclin B1 and Cdc2 protein levels (peaking between 12 and 24 h post treatment), and their levels started to decline after the initial increase. This early up-regulation of cyclin B1 and Cdc2 closely matched in timing the nocodazole-induced mitotic prometaphase arrest. Selective knockdown of cyclin B1or Cdc2 each abrogated nocodazole-induced accumulation of prometaphase cells. The nocodazole-induced prometaphase arrest was also abrogated by pre-treatment of cells with roscovitine, an inhibitor of cyclin-dependent kinases, or with cycloheximide, a protein synthesis inhibitor that was found to suppress cyclin B1 and Cdc2 up-regulation. In addition, we found that MAD2 knockdown abrogated nocodazole-induced accumulation of cyclin B1 and Cdc2 proteins, which was accompanied by an attenuation of nocodazole-induced prometaphase arrest. CONCLUSIONS/SIGNIFICANCE: These observations demonstrate that the strong early up-regulation of cyclin B1 and Cdc2 contributes critically to the rapid and selective accumulation of prometaphase-arrested cells, a phenomenon associated with exposure to microtubule inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Cyclin B1/metabolism , Cyclin B/metabolism , Mitosis/drug effects , Nocodazole/pharmacology , Prometaphase/drug effects , Blotting, Western , Breast Neoplasms/pathology , CDC2 Protein Kinase , Cell Line, Tumor , Cyclin-Dependent Kinases , Female , Flow Cytometry , Humans , Microscopy, Fluorescence , RNA, Small Interfering
13.
Br J Pharmacol ; 162(1): 175-92, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20955365

ABSTRACT

BACKGROUND AND PURPOSE: Glutamate-induced oxidative stress plays a critical role in the induction of neuronal cell death in a number of disease states. We sought to determine the role of the c-Jun NH(2) -terminal kinase (JNK)-p53-growth arrest and DNA damage-inducible gene (GADD) 45α apoptotic cascade in mediating glutamate-induced oxidative cytotoxicity in hippocampal neuronal cells. EXPERIMENTAL APPROACH: HT22 cells, a mouse hippocampal neuronal cell line, were treated with glutamate to induce oxidative stress in vitro. Kainic acid-induced oxidative damage to the hippocampus in rats was used as an in vivo model. The signalling molecules along the JNK-p53-GADD45α cascade were probed with various means to determine their contributions to oxidative neurotoxicity. KEY RESULTS: Treatment of HT22 cells with glutamate increased the mRNA and protein levels of GADD45α, and these increases were suppressed by p53 knock-down. Knock-down of either p53 or GADD45α also prevented glutamate-induced cell death. Glutamate-induced p53 activation was preceded by accumulation of reactive oxygen species, and co-treatment with N-acetyl-cysteine prevented glutamate-induced p53 activation and GADD45α expression. Knock-down of MKK4 or JNK, or the presence of SP600125 (a JNK inhibitor), each inhibited glutamate-induced p53 activation and GADD45α expression. In addition, we also confirmed the involvement of GADD45α in mediating kainic acid-induced hippocampal oxidative neurotoxicity in vivo. CONCLUSIONS: AND IMPLICATIONS Activation of the JNK-p53-GADD45α cascade played a critical role in mediating oxidative cytotoxicity in hippocampal neurons. Pharmacological inhibition of this signalling cascade may provide an effective strategy for neuroprotection.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , Hippocampus/metabolism , MAP Kinase Kinase 4/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Oxidative Stress , Tumor Suppressor Protein p53/metabolism , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Cell Line , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/enzymology , Immunohistochemistry , In Situ Nick-End Labeling , Kainic Acid/pharmacology , Mice , Microscopy, Fluorescence , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Nuclear Proteins/genetics , Polymerase Chain Reaction , RNA, Small Interfering , Rats , Reactive Oxygen Species/metabolism
14.
PLoS One ; 5(8): e11951, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20700524

ABSTRACT

BACKGROUND: A combination of levodopa (L-DOPA) and carbidopa is the most commonly-used treatment for symptom management in Parkinson's disease. Studies have shown that concomitant use of a COMT inhibitor is highly beneficial in controlling the wearing-off phenomenon by improving L-DOPA bioavailability as well as brain entry. The present study sought to determine whether (-)-epigallocatechin-3-gallate (EGCG), a common tea polyphenol, can serve as a naturally-occurring COMT inhibitor that also possesses neuroprotective actions. METHODOLOGY/PRINCIPAL FINDINGS: Using both in vitro and in vivo models, we investigated the modulating effects of EGCG on L-DOPA methylation as well as on chemically induced oxidative neuronal damage and degeneration. EGCG strongly inhibited human liver COMT-mediated O-methylation of L-DOPA in a concentration-dependent manner in vitro, with an average IC50 of 0.36 microM. Oral administration of EGCG moderately lowered the accumulation of 3-O-methyldopa in the plasma and striatum of rats treated with L-DOPA+carbidopa. In addition, EGCG also reduced glutamate-induced oxidative cytotoxicity in cultured HT22 mouse hippocampal neuronal cells through inactivation of the nuclear factor kappaB-signaling pathway. Under in vivo conditions, administration of EGCG exerted a strong protective effect against kainic acid-induced oxidative neuronal death in the hippocampus of rats. CONCLUSIONS/SIGNIFICANCE: These observations suggest that oral administration of EGCG may have significant beneficial effects in Parkinson's patients treated with L-DOPA and carbidopa by exerting a modest inhibition of L-DOPA methylation plus a strong neuroprotection against oxidative damage and degeneration.


Subject(s)
Biological Products/pharmacology , Catechin/analogs & derivatives , Enzyme Inhibitors/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Levodopa/metabolism , Animals , Catechin/pharmacology , Catechol O-Methyltransferase Inhibitors , Cell Death/drug effects , Cell Line , Hippocampus/metabolism , Humans , Kainic Acid/pharmacology , Male , Methylation/drug effects , Mice , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley
15.
Free Radic Biol Med ; 49(5): 800-13, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20542495

ABSTRACT

Oxidative stress can induce cytotoxicity in neurons, which plays an important role in the etiology of neuronal damage and degeneration. This study sought to determine the cellular and biochemical mechanisms underlying resveratrol's protective effect against oxidative neuronal death. Cultured HT22 cells, an immortalized mouse hippocampal neuronal cell line, were used as an in vitro model, and oxidative stress and neurotoxicity were induced in these neuronal cells by exposure to high concentrations of glutamate. Resveratrol strongly protected HT22 cells from glutamate-induced oxidative cell death. Resveratrol's neuroprotective effect was independent of its direct radical scavenging property, but instead was dependent on its ability to selectively induce the expression of mitochondrial superoxide dismutase (SOD2) and, subsequently, reduce mitochondrial oxidative stress and damage. The induction of mitochondrial SOD2 by resveratrol was mediated through the activation of the PI3K/Akt and GSK-3beta/beta-catenin signaling pathways. Taken together, the results of this study show that up-regulation of mitochondrial SOD2 by resveratrol represents an important mechanism for its protection of neuronal cells against oxidative cytotoxicity resulting from mitochondrial oxidative stress.


Subject(s)
Cytoprotection/drug effects , Neurons/drug effects , Oxidative Stress/drug effects , Stilbenes/pharmacology , Animals , Antioxidants/pharmacology , Cell Death/drug effects , Cell Line , Drug Evaluation, Preclinical , Glutamic Acid/toxicity , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiology , Mice , Neurons/metabolism , Neurons/physiology , Neuroprotective Agents/pharmacology , Oxidative Stress/physiology , Phosphatidylinositol 3-Kinases/metabolism , Reactive Oxygen Species/metabolism , Resveratrol , Signal Transduction/drug effects , Superoxide Dismutase/metabolism
16.
Mol Carcinog ; 49(8): 750-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20572158

ABSTRACT

Earlier studies have shown that resveratrol could induce death in several human cancer cell lines in culture. Here we report our observation that resveratrol can also promote the growth of certain human cancer cells when they are grown either in culture or in athymic nude mice as xenografts. At relatively low concentrations (

Subject(s)
NF-kappa B/metabolism , Phenols/pharmacology , Stilbenes/pharmacology , Animals , Female , Humans , Mice , Mice, Nude , NF-kappa B/genetics , NF-kappa B/pharmacology , Neoplasms/genetics , Nervous System Neoplasms/genetics , Nitriles , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Resveratrol , Signal Transduction/drug effects , Signal Transduction/genetics , Sulfones
17.
PLoS One ; 5(4): e10296, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20421971

ABSTRACT

BACKGROUND: The present study sought to further investigate the in vitro and in vivo anticancer effects of a representative omega-3 fatty acid, docosahexaenoic acid (DHA), with a focus on assessing the induction of oxidative stress and apoptosis as an important mechanism for its anticancer actions. METHODOLOGY/PRINCIPAL FINDINGS: In vitro studies showed that DHA strongly reduces the viability and DNA synthesis of MCF-7 human breast cancer cells in culture, and also promotes cell death via apoptosis. Mechanistically, accumulation of reactive oxygen species and activation of caspase 8 contribute critically to the induction of apoptotic cell death. Co-presence of antioxidants or selective inhibition or knockdown of caspase 8 each effectively abrogates the cytotoxic effect of DHA. Using athymic nude mice as an in vivo model, we found that feeding animals the 5% fish oil-supplemented diet for 6 weeks significantly reduces the growth of MCF-7 human breast cancer cells in vivo through inhibition of cancer cell proliferation as well as promotion of cell death. Using 3-nitrotyrosine as a parameter, we confirmed that the fish oil-supplemented diet significantly increases oxidative stress in tumor cells in vivo. Analysis of fatty acid content in plasma and tissues showed that feeding animals a 5% fish oil diet increases the levels of DHA and eicosapentaenoic acid in both normal and tumorous mammary tissues by 329% and 300%, respectively. CONCLUSIONS/SIGNIFICANCE: DHA can strongly induce apoptosis in human MCF-7 breast cancer cells both in vitro and in vivo. The induction of apoptosis in these cells is selectively mediated via caspase 8 activation. These observations call for further studies to assess the effectiveness of fish oil as a dietary supplement in the prevention and treatment of human breast cancer.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Caspase 8/metabolism , Docosahexaenoic Acids/pharmacology , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Fatty Acids, Omega-3/pharmacology , Female , Fish Oils/administration & dosage , Fish Oils/therapeutic use , Humans , Mice , Mice, Nude , Oxidative Stress , Xenograft Model Antitumor Assays
18.
Eur J Cancer ; 46(10): 1882-91, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20223651

ABSTRACT

It was reported recently that resveratrol could sensitise a number of cancer cell lines to the anticancer actions of several other cancer drugs, including paclitaxel. In the present study, we further investigated whether resveratrol could sensitise human breast cancer cells to paclitaxel-induced cell death. Unexpectedly, we found that resveratrol strongly diminished the susceptibility of MDA-MB-435s, MDA-MB-231 and SKBR-3 cells to paclitaxel-induced cell death in culture, although this effect was not observed in MCF-7 cells. Using MDA-MB-435s cells as a representative model, a similar observation was made in athymic nude mice. Mechanistically, the modulating effect of resveratrol was partially attributable to its inhibition of paclitaxel-induced G(2)/M cell cycle arrest, together with an accumulation of cells in the S-phase. In addition, resveratrol could suppress paclitaxel-induced accumulation of reactive oxygen species (ROS) and subsequently the inactivation of anti-apoptotic Bcl-2 family proteins. These observations suggest that the strategy of concomitant use of resveratrol with paclitaxel is detrimental in certain types of human cancers. Given the widespread use of resveratrol among cancer patients, this study calls for more preclinical and clinical testing of the potential benefits and harms of using resveratrol as a dietary adjuvant in cancer patients.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Breast Neoplasms/drug therapy , Paclitaxel/antagonists & inhibitors , Stilbenes/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Resveratrol , Transplantation, Heterologous , bcl-X Protein/metabolism
19.
Free Radic Biol Med ; 48(6): 821-30, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20060889

ABSTRACT

Oxidative cell death is an important contributing factor in neurodegenerative diseases. Using HT22 mouse hippocampal neuronal cells as a model, we sought to demonstrate that mitochondria are crucial early targets of glutamate-induced oxidative cell death. We show that when HT22 cells were transfected with shRNA for knockdown of the mitochondrial superoxide dismutase (SOD2), these cells became more susceptible to glutamate-induced oxidative cell death. The increased susceptibility was accompanied by increased accumulation of mitochondrial superoxide and loss of normal mitochondrial morphology and function at early time points after glutamate exposure. However, overexpression of SOD2 in these cells reduced the mitochondrial superoxide level, protected mitochondrial morphology and functions, and provided resistance against glutamate-induced oxidative cytotoxicity. The change in the sensitivity of these SOD2-altered HT22 cells was neurotoxicant-specific, because the cytotoxicity of hydrogen peroxide was not altered in these cells. In addition, selective knockdown of the cytosolic SOD1 in cultured HT22 cells did not appreciably alter their susceptibility to either glutamate or hydrogen peroxide. These findings show that the mitochondrial SOD2 plays a critical role in protecting neuronal cells from glutamate-induced oxidative stress and cytotoxicity. These data also indicate that mitochondria are important early targets of glutamate-induced oxidative neurotoxicity.


Subject(s)
Mitochondria/enzymology , Neurons/metabolism , Superoxide Dismutase/metabolism , Animals , Cell Death/drug effects , Cells, Cultured , Glutamic Acid/toxicity , Mice , Neurons/drug effects , Neurons/enzymology , Oxidative Stress/drug effects
20.
Br J Pharmacol ; 158(2): 462-74, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19563536

ABSTRACT

BACKGROUND AND PURPOSE: Resveratrol (RES) has been shown to prolong lifespan and prevent cancer formation. At present, the precise cellular mechanisms of RES actions are still not clearly understood, and this is the focus of this study. EXPERIMENTAL APPROACH: Using human hepatocellular carcinoma-derived HepG2 cells as a model, we studied RES-induced changes in cell growth, cell cycle progression and apoptosis. KEY RESULTS: RES at lower concentrations induced a strong but reversible S-phase delay and mild DNA synthesis inhibition, yet without causing apoptotic or necrotic cell death. At high concentrations, RES induced apoptosis, which is mainly mediated by the mitochondrial pathway. Overall, RES was a relatively weak apoptotic agent. Mechanistically, MEK inhibition was identified as an important early signalling event for RES-induced apoptosis. In comparison, activation of CDK2 and checkpoint kinase 2, and inhibition of phosphatidylinositol 3'-kinase/Akt signalling pathway contributed to the induction by RES of a reversible, non-cytotoxic S-phase delay. CONCLUSION AND IMPLICATIONS: It is hypothesized that the induction of a non-cytotoxic S-phase delay may represent a useful mechanistic strategy for lifespan prolongation and cancer prevention. When cell cycles are selectively slowed down in the S phase, it would cumulatively increase the total lifespan of an organism if the total numbers of cell divisions of a given organism are assumed to remain basically constant. Likewise, when cells proceed through the cell cycles at a reduced pace during DNA replication, it may allow cells more time to repair the damaged DNA, and thereby reduce the chances for mutagenesis and tumour initiation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Stilbenes/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA/biosynthesis , DNA/drug effects , Dose-Response Relationship, Drug , Humans , Membrane Potential, Mitochondrial/drug effects , Resveratrol , S Phase/drug effects , Signal Transduction/drug effects , Stilbenes/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...