Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Dig Dis Sci ; 64(12): 3528-3538, 2019 12.
Article in English | MEDLINE | ID: mdl-31273592

ABSTRACT

BACKGROUND: α-Hederin has been shown promising anti-tumor potential against various cancer cell lines. However, reports about effects of α-hederin on esophageal squamous cell carcinoma (ESCC) are still unavailable. AIM: To investigate the inhibitory effects of α-hederin on ESCC and explore the underlying mechanism. METHODS: Human esophageal carcinoma cell line (Eca-109) was used for the experiment. Cell Counting Kit-8, flow cytometry, Hoechst 33258 staining, enhanced ATP assay kit, 2',7'-dichlorofluorescin diacetate, JC-1 kit, and Western bolt were used to assess the cell viability, cycle, apoptosis, cellular ATP content, reactive oxygen species (ROS) level, mitochondrial membrane potential (MMP), and protein expression, respectively, in vitro. Xenografted tumor model was constructed to evaluate the in vivo anti-tumor effects of α-hederin. RESULTS: Compared with control group, α-hederin significantly inhibited the proliferation, induced apoptosis of ESCC, and arrested the cell cycle in G1 phase (P < 0.05). α-Hederin induced the accumulation of ROS, decrement of ATP levels, and disruption of MMP (P < 0.05). The detection of mitochondrial and cytosol proteins showed that AIF, Apaf-1, and Cyt C were released and increased in cytoplasm, and then, caspase-3, caspase-9, and Bax were involved and increased, while Bcl-2 level was decreased (P < 0.05). Furthermore, the above changes were amplified in the group pretreated with L-buthionine sulfoximine, while N-acetyl-L-cysteine plays an opposite role (P < 0.05). Meanwhile, α-hederin significantly inhibited the growth of xenografted tumors with favorable safety. CONCLUSION: α-Hederin could inhibit the proliferation and induce apoptosis of ESCC via dissipation of the MMP with simultaneous ROS generation and activation of the mitochondrial pathway.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Oleanolic Acid/analogs & derivatives , Reactive Oxygen Species/metabolism , Saponins/pharmacology , Adenosine Triphosphate/metabolism , Animals , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Apoptotic Protease-Activating Factor 1/drug effects , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Caspase 9/drug effects , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D1/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytochromes c/drug effects , Cytochromes c/metabolism , Flow Cytometry , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Male , Mice, Nude , Mitochondria/metabolism , Neoplasm Transplantation , Oleanolic Acid/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
2.
J Cell Physiol ; 233(4): 3638-3647, 2018 04.
Article in English | MEDLINE | ID: mdl-29030979

ABSTRACT

Pioglitazone is an anti-diabetic agent that belongs to the thiazolidinedione class, which target peroxisome proliferator-activated receptor γ (PPARγ), a transcription factor in the nuclear receptor family. Different cancer cells expressing high levels of PPARγ and PPARγ ligands induce cell cycle arrest, cell differentiation, and apoptosis. However, the mechanisms underlying these processes remain unknown. Here, we investigated the mechanism underlying pioglitazone-induced apoptosis in human cancer cells. We showed that at similar concentrations, pioglitazone induced death in cancer cells expressing high or low levels of PPARγ. Combined treatment of pioglitazone and GW9662, a PPARγ antagonist, did not rescue this cell death phenotype. Z-VAD-fmk, a pan-caspase inhibitor, did not reverse pioglitazone-induced apoptosis in cancer cells expressing PPARγ at high or low levels. Pioglitazone suppressed the activation of signal transducers and activator of transcription 3 (STAT3) and Survivin expression, and enhanced the apoptosis-inducing factor (AIF) levels in these cells. Furthermore, pioglitazone enhanced the cytotoxic effect of cisplatin and oxaliplatin by suppressing Survivin and increasing AIF expression. These results indicated that pioglitazone induced apoptosis via a PPARγ-independent pathway, thus describing pioglitazone as a potential therapeutic agent for controlling the progression of different cancers.


Subject(s)
PPAR gamma/drug effects , Pioglitazone/pharmacology , STAT3 Transcription Factor/drug effects , Signal Transduction/drug effects , Apoptosis/drug effects , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Cell Line, Tumor , Humans , Hypoglycemic Agents/pharmacology , Inhibitor of Apoptosis Proteins/metabolism , PPAR gamma/metabolism , STAT3 Transcription Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factors/metabolism
3.
Am J Physiol Heart Circ Physiol ; 310(3): H376-84, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26637561

ABSTRACT

Calpain 1 (CPN1) is a ubiquitous cysteine protease that exists in both cytosol and cardiac mitochondria. Mitochondrial CPN1 (mit-CPN1) is located in the intermembrane space and matrix. Activation of mit-CPN1 within the intermembrane space increases cardiac injury by releasing apoptosis-inducing factor from mitochondria during ischemia-reperfusion (IR). We asked if activation of mit-CPN1 is involved in mitochondrial injury during IR. MDL-28170 (MDL) was used to inhibit CPN1 in buffer-perfused hearts following 25-min ischemia and 30-min reperfusion. MDL treatment decreased the release of lactate dehydrogenase into coronary effluent compared with untreated hearts, indicating that inhibition of CPN1 decreases cardiac injury. MDL also prevented the cleavage of spectrin (a substrate of CPN1) in cytosol during IR, supporting that MDL treatment decreased cytosolic calpain activation. In addition, MDL markedly improved calcium retention capacity compared with untreated heart, suggesting that MDL treatment decreases mitochondrial permeability transition pore opening. In addition, we found that IR led to decreased complex I activity, whereas inhibition of mit-CPN1 using MDL protected complex I. Pyruvate dehydrogenase content was decreased following IR. However, pyruvate dehydrogenase content was preserved in MDL-treated mitochondria. Taken together, MDL treatment decreased cardiac injury during IR by inhibiting both cytosolic and mit-CPN1. Activation of mit-CPN1 increases cardiac injury during IR by sensitizing mitochondrial permeability transition pore opening and impairing mitochondrial metabolism through damage of complex I.


Subject(s)
Apoptosis Inducing Factor/drug effects , Calpain/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Heart/drug effects , Mitochondria, Heart/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Myocardial Reperfusion Injury/metabolism , Animals , Apoptosis Inducing Factor/metabolism , Calcium/metabolism , Calpain/metabolism , Cytosol/drug effects , Cytosol/metabolism , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Spectrin/drug effects , Spectrin/metabolism
4.
J Cardiothorac Vasc Anesth ; 29(5): 1172-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26205644

ABSTRACT

OBJECTIVES: To compare the effects of propofol, sevoflurane, and the combination of the 2 on circulating lymphocytes in patients undergoing off-pump coronary artery bypass graft (OPCAB) surgery. DESIGN: A prospective, randomized study. SETTING: A university hospital. PARTICIPANTS: One hundred five patients undergoing elective OPCAB surgery. INTERVENTIONS: Participants were randomized to receive sevoflurane (group S), propofol (group P), or coadministration (group C) of sevoflurane- and propofol-maintained anesthesia. MEASUREMENTS AND MAIN RESULTS: Blood samples were obtained before, during, and after surgery. Caspase-3 and apoptosis-inducing factor in lymphocytes were evaluated by Western blot. During surgery, 5 minutes after revascularization of the left anterior descending artery, 5 minutes after all anastomoses (T4), and after the sternal closure (T5), caspase-3 expression of group S was higher than that of group P (p = 0.02) and group C (p = 0.02). At T4 and T5, expression of active apoptosis-inducing factor in group S was higher than that in the other 2 groups (p = 0.03 and p = 0.04, respectively). 24 hours after surgery, the lymphocyte count of group S (0.55/nL) was lower than that of group P (0.73/nL, p = 0.02) and group C (0.73/nL, p = 0.03). Intensive care unit stay of group S (3.0 days) was longer than that of the other 2 groups (2.2 days, p = 0.02 and 2.1 days, p = 0.01). CONCLUSIONS: OPCAB surgery was associated with postoperative lymphopenia. Regarding a protective effect for circulating lymphocytes, propofol and the combination of sevoflurane- and propofol-maintained anesthesia were both superior to sevoflurane-maintained anesthesia.


Subject(s)
Anesthetics, Inhalation/pharmacology , Anesthetics, Intravenous/pharmacology , Coronary Artery Bypass, Off-Pump , Lymphocytes/drug effects , Methyl Ethers/pharmacology , Propofol/pharmacology , Adolescent , Adult , Aged , Aged, 80 and over , Anesthetics, Inhalation/blood , Anesthetics, Intravenous/blood , Apoptosis Inducing Factor/blood , Apoptosis Inducing Factor/drug effects , Blotting, Western , Caspase 3/blood , Caspase 3/drug effects , Drug Therapy, Combination , Humans , Male , Methyl Ethers/blood , Middle Aged , Propofol/blood , Prospective Studies , Sevoflurane , Young Adult
5.
Int J Clin Exp Pathol ; 8(10): 12207-15, 2015.
Article in English | MEDLINE | ID: mdl-26722405

ABSTRACT

Poly(ADP-ribose)polymerase-1 (PARP-1) is anubiquitous, DNA repair-associated enzyme, which participates in gene expression, cell death, central nerve system (CNS) disorders and oxidative stress. According to the previous studies, PARP-1 over-activation may lead to over-consumption of ATP and even cell apoptosis. Spinal cord injury (SCI) is an inducement towards PARP-1 over-activation due to its massive damage to DNA. 3-aminobenzamide (3-AB) is a kind of PARP-1 inhibitors. The relationship among PARP-1, 3-AB, SCI and apoptosis has not been fully understood. Hence, in the present study, we focused on the effects of 3-AB on cell apoptosis after SCI. Accordingly, SCI model was constructed artificially, and 3-AB was injected intrathecally into the Sprague-Dawley (SD) rats. The results demonstrated an increase in cell apoptosis after SCI. Furthermore, PARP-1 was over-activated after SCI but inhibited by 3-AB injection. In addition, apoptosis-inducing factor (AIF) was inhibited but B-cell lymphoma-2 (Bcl-2) was up-regulated by 3-AB. Interestingly, caspase-3 was not significantly altered with or without 3-AB. In conclusion, our experiments showed that 3-AB, as a PARP-1 inhibitor, could inhibit cell apoptosis after SCI in caspase-independent way, which could provide a better therapeutic target for the treatment of SCI.


Subject(s)
Apoptosis/drug effects , Benzamides/pharmacology , Poly (ADP-Ribose) Polymerase-1/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/genetics , Apoptosis Inducing Factor/metabolism , Caspase 3/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Disease Models, Animal , Humans , Injections, Spinal , Male , Oxidative Stress/drug effects , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley
6.
ChemMedChem ; 7(5): 805-14, 2012 May.
Article in English | MEDLINE | ID: mdl-22383263

ABSTRACT

Fourteen silver(I) complexes bearing N-heterocyclic carbene (NHC) ligands were prepared and evaluated for anticancer activity. Some of these were found to exhibit potent antiproliferative activity toward several types of human cancer cell lines, including drug-resistant cell lines, with IC(50) values in the nanomolar range. An initial investigation into the mechanism of cell death induced by this family of silver(I) complexes was carried out. Cell death was shown to result from the activation of apoptosis without involvement of primary necrosis. In HL60 cells, silver-NHCs induce depolarization of the mitochondrial membrane potential (ΔΨ(m)) and likely allow the release of mitochondrial proteins to elicit early apoptosis. This effect is not related to the overproduction of reactive oxygen species (ROS). In addition, apoptosis is not associated with the activation of caspase-3, but is triggered by the translocation of apoptosis-inducing factor (AIF) and caspase-12 from mitochondria and the endoplasmic reticulum, respectively, into the nucleus to promote DNA fragmentation and ultimately cell death. No modification in cell-cycle distribution was observed, indicating that silver-NHCs are not genotoxic. Finally, the use of a fluorescent complex showed that silver-NHCs target mitochondria. Altogether, these results demonstrate that silver-NHCs induce cancer cell death independent of the caspase cascade via the mitochondrial AIF pathway.


Subject(s)
Coordination Complexes , Heterocyclic Compounds , Methane/analogs & derivatives , Silver , Apoptosis/drug effects , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Drug Resistance, Neoplasm , Flow Cytometry , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Humans , Inhibitory Concentration 50 , Methane/chemistry , Methane/pharmacology , Models, Biological , Molecular Structure , Silver/pharmacology
7.
Mar Drugs ; 9(4): 603-614, 2011.
Article in English | MEDLINE | ID: mdl-21731552

ABSTRACT

Fucan is a term used to denominate a family of sulfated polysaccharides rich in sulfated l-fucose. Heterofucan SF-1.5v was extracted from the brown seaweed Sargassum filipendula by proteolytic digestion followed by sequential acetone precipitation. This fucan showed antiproliferative activity on Hela cells and induced apoptosis. However, SF-1.5v was not able to activate caspases. Moreover, SF-1.5v induced glycogen synthase kinase (GSK) activation, but this protein is not involved in the heterofucan SF-1.5v induced apoptosis mechanism. In addition, ERK, p38, p53, pAKT and NFκB were not affected by the presence of SF-1.5v. We determined that SF-1.5v induces apoptosis in HeLa mainly by mitochondrial release of apoptosis-inducing factor (AIF) into cytosol. In addition, SF-1.5v decreases the expression of anti-apoptotic protein Bcl-2 and increased expression of apoptogenic protein Bax. These results are significant in that they provide a mechanistic framework for further exploring the use of SF-1.5v as a novel chemotherapeutics against human cervical cancer.


Subject(s)
Apoptosis/drug effects , Fucose/chemistry , Polysaccharides/pharmacology , Sargassum/chemistry , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Cell Proliferation/drug effects , Cytosol/drug effects , Cytosol/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Polysaccharides/isolation & purification , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology
8.
Arch Pharm Res ; 33(8): 1181-91, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20803121

ABSTRACT

Dietary polyphenols have been correlated with a reduced risk of developing cancer. Quercetin (a natural polyphenolic compound) induced apoptosis in many human cancer cell lines, including breast cancer MCF-7 cells. However, the involvement of possible signaling pathways and the roles of quercetin in apoptosis are still undefined. The purpose of this study was to investigate the effects of quercetin on the induction of the apoptotic pathway in human breast cancer MCF-7 cells. When MCF-7 cells were treated with quercetin for 24 and 48 h and at various doses (10-175 microM), cell viability decreased significantly in time- and dose-dependent manners. Exposure of MCF-7 cells to 10-175 microM quercetin resulted in an approximate 90.25% decrease in viable cells. To explicate the mechanism underlying the antiproliferative effect of quercetin, cell cycle distribution and apoptosis in MCF-7 cells was investigated after exposure to 150 microM quercetin for 6-48 h. Quercetin caused a remarkable increase in the number of S phase (14.56% to 61.35%) and sub-G1 phase cells (0.1% to 8.32%) in a dose- and time-dependent manner. Quercetin caused S phase arrest by decreasing the protein expression of CDK2, cyclins A and B while increasing the p53 and p57 proteins. Following incubation with quercetin for 48 h, MCF-7 cells showed apoptotic cell death by the decreased levels of Bcl-2 protein and DeltaPsi(m) and increased activations of caspase-6, -8 and -9. Moreover, quercetin increased the AIF protein released from mitochondria to nuclei and the GADD153 protein translocation from endoplasmic reticulum to the nuclei. These data suggested that quercetin may induce apoptosis by direct activation of the caspase cascade through the mitochondrial pathway in MCF-7 cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Quercetin/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Breast Neoplasms/pathology , Caspases/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Dose-Response Relationship, Drug , Female , Humans , Mitochondria/drug effects , Quercetin/administration & dosage , Time Factors , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/metabolism
9.
J Dent Res ; 89(7): 689-94, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20439934

ABSTRACT

Reactive oxygen species (ROS) are essential for the induction of T-cell apoptosis by butyric acid, an extracellular metabolite of periodontopathic bacteria. To determine the involvement of oxidative stress in apoptosis pathways, we investigated the contribution of ROS in mitochondrial signaling pathways, death-receptor-initiated signaling pathway, and endoplasmic reticulum stress in butyric-acid-induced T-cell apoptosis. N-acetyl-L-Cysteine (NAC) abrogated mitochondrial injury, cytochrome c, AIF, and Smac release, and Bcl-2 and Bcl-xL suppression and Bax and Bad activation induced by butyric acid. However, the decrease in cFLIP expression by butyric acid was not restored by treatment with NAC; increases in caspase-4 and -10 activities by butyric acid were completely abrogated by NAC. NAC also affected the elevation of GRP78 and CHOP/GADD153 expression by butyric acid. These results suggest that butyric acid is involved in mitochondrial-dysfunction- and endoplasmic reticulum stress-mediated apoptosis in human Jurkat T-cells via a ROS-dependent mechanism.


Subject(s)
Apoptosis/drug effects , Butyric Acid/pharmacology , Oxidative Stress/physiology , T-Lymphocytes/drug effects , Acetylcysteine/pharmacology , Apoptosis Inducing Factor/drug effects , Apoptosis Regulatory Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein/drug effects , Caspase 10/drug effects , Caspase Inhibitors , Caspases, Initiator/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Cytochromes c/drug effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Free Radical Scavengers/pharmacology , Heat-Shock Proteins/drug effects , Humans , Inhibitor of Apoptosis Proteins/pharmacology , Intracellular Signaling Peptides and Proteins/pharmacology , Jurkat Cells , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondrial Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Reactive Oxygen Species/pharmacology , Receptors, Death Domain/drug effects , Serpins/pharmacology , Signal Transduction/drug effects , Viral Proteins/pharmacology , bcl-2-Associated X Protein/drug effects , bcl-Associated Death Protein/drug effects , bcl-X Protein/drug effects
10.
J Neurosci ; 30(8): 2967-78, 2010 Feb 24.
Article in English | MEDLINE | ID: mdl-20181594

ABSTRACT

Poly(ADP-ribose)-1 (PARP-1) is a key mediator of cell death in excitotoxicity, ischemia, and oxidative stress. PARP-1 activation leads to cytosolic NAD(+) depletion and mitochondrial release of apoptosis-inducing factor (AIF), but the causal relationships between these two events have been difficult to resolve. Here, we examined this issue by using extracellular NAD(+) to restore neuronal NAD(+) levels after PARP-1 activation. Exogenous NAD(+) was found to enter neurons through P2X(7)-gated channels. Restoration of cytosolic NAD(+) by this means prevented the glycolytic inhibition, mitochondrial failure, AIF translocation, and neuron death that otherwise results from extensive PARP-1 activation. Bypassing the glycolytic inhibition with the metabolic substrates pyruvate, acetoacetate, or hydroxybutyrate also prevented mitochondrial failure and neuron death. Conversely, depletion of cytosolic NAD(+) with NAD(+) glycohydrolase produced a block in glycolysis inhibition, mitochondrial depolarization, AIF translocation, and neuron death, independent of PARP-1 activation. These results establish NAD(+) depletion as a causal event in PARP-1-mediated cell death and place NAD(+) depletion and glycolytic failure upstream of mitochondrial AIF release.


Subject(s)
NAD/deficiency , Nerve Degeneration/enzymology , Nerve Degeneration/physiopathology , Neurons/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Animals , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Cell Death/physiology , Cell Respiration/drug effects , Cell Respiration/physiology , Cells, Cultured , Energy Metabolism/drug effects , Energy Metabolism/physiology , Glycolysis/drug effects , Glycolysis/physiology , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/enzymology , Mitochondrial Diseases/physiopathology , NAD/pharmacology , Neurons/drug effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Protein Transport/drug effects , Protein Transport/physiology , Receptors, Purinergic P2/drug effects , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2X7
11.
Cancer Lett ; 290(1): 68-75, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-19781850

ABSTRACT

(-)-Epigallocatechin-3-gallate (EGCG), a major green tea polyphenol, was tested for in vitro cytotoxicity against human laryngeal epidermoid carcinoma of the larynx Hep2 cells. EGCG-induced apoptotic cell death accompanied by a change in the cell cycle. However, EGCG did not result in caspase activation, nor did a caspase inhibitor block cell death. Furthermore, EGCG caused no change in the intracellular levels of reactive oxygen species (ROS). The levels of p53 were increased in the EGCG-treated cells, with a corresponding decrease in Bcl-2 and Bid protein levels as well as an increase in the Bax level. In addition, EGCG induced the cytoplasmic release of cytochrome c from the mitochondria accompanied by a decreased mitochondrial membrane potential, and subsequently upregulated translocation of apoptosis-inducing factor (AIF) and endonuclease G (EndoG) into the nucleus during the apoptotic process. Taken together, these findings indicate that the p53-mediated mitochondrial pathway and the nuclear translocation of AIF and EndoG play a crucial role in EGCG-induced apoptosis of human laryngeal epidermoid carcinoma Hep2 cells, which proceeds through a caspase-independent pathway.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis Inducing Factor/metabolism , Carcinoma, Squamous Cell/metabolism , Catechin/analogs & derivatives , Endodeoxyribonucleases/metabolism , Laryngeal Neoplasms/metabolism , Apoptosis Inducing Factor/drug effects , Blotting, Western , Catechin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Endodeoxyribonucleases/drug effects , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism
12.
J Periodontal Res ; 44(6): 767-75, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19602127

ABSTRACT

BACKGROUND AND OBJECTIVE: We reported previously that cyclosporine A induces a high level of expression of p21 in rat gingival keratinocytes and in OECM1 cells. In this study, the apoptosis of gingival keratinocytes after treatment with cyclosporine A was evaluated using the same models. MATERIAL AND METHODS: Forty Sprague-Dawley rats with right edentulous ridges were assigned into cyclosporine A (30 mg/kg) and control groups. Four weeks later, gingivae were screened for expression of apoptotic genes using microarray analyses and DNA fragmentation. The expression of bcl2-associated X protein (Bax), apoptosis-inducing factor (AIF) and Caspase 3 mRNAs, and the expression of Bax, AIF, Caspase 9 and Fas proteins, were analyzed using the reverse transcription-polymerase chain reaction and immunohistochemistry, respectively. Apoptosis in OECM1 cells (keratinocytes of a gingival carcinoma cell line), after treatment with cyclosporine A, was evaluated by 4',6-diamidino-2-phenylindole (DAPI) staining and flow cytometry, whereas the expression of Bax, AIF, Caspase 3 and 8, Bcl-2 and Fas proteins were examined using western blotting. RESULTS: According to microarray analyses, the expression of certain apoptotic genes was altered in the gingiva of rats who received cyclosporine A, and increased number of DNA fragments were detected. Expression of mRNA or protein for Bax, AIF and Caspase 3 and 9 in the gingivae of rats increased after treatment with cyclosporine A. An increased number of apoptotic bodies and of OECM1 cells in the sub-G1 phase was observed after treatment with cyclosporine A. Increased expression of AIF, Bax and Caspase 3 protein, but not of bcl-2, Caspase 8 or Fas protein, was observed in cells after treatment with cyclosporine A. CONCLUSION: Based on the above findings, we suggest that cyclosporine A might enhance the apoptosis of gingival keratinocytes, mainly via the mitochondrial pathway.


Subject(s)
Apoptosis/drug effects , Cyclosporine/pharmacology , Gingiva/drug effects , Immunosuppressive Agents/pharmacology , Keratinocytes/drug effects , Mitochondria/drug effects , Animals , Apoptosis Inducing Factor/drug effects , Carcinoma, Squamous Cell/pathology , Caspase 3/drug effects , Caspase 8/drug effects , Caspase 9/drug effects , Cell Line, Tumor , DNA Fragmentation , Flow Cytometry , Fluorescent Dyes , Gingiva/cytology , Humans , Indoles , Male , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-bcl-2/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley , bcl-2-Associated X Protein/drug effects , fas Receptor/drug effects
13.
Cell Biol Int ; 33(9): 964-70, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19573614

ABSTRACT

In our previous studies, we demonstrated that the deglycosylation of bleomycin-A2 (BLM-A2) does not affect the capacity of this drug to induce cell death by apoptosis in a caspase-independent manner in laryngeal cancer cells (HEp-2), but suppresses the ability of BLM-A2 to induce ROS formation. We have now investigated the consequence of BLM-A2 deglycosylation in terms of the involvement of apoptotic pathways in HEp-2 cells. Apoptosis induced by bleomycin-A2 and deglyco-BLM-A2 is associated with the release of cytochrome c and AIF. Only Bax was oligomerized with BLM-A2-induced HEp-2 cell death. BLM-A2 and deglyco-BLM-A2-induced apoptosis depended on JNK activation but was independent of death receptors expression. In contrast, both of these drugs would sensitize HEp-2 cells to death receptor ligand-induced cell death. These observations indicate that the deglycosylation of BLM does not impair the ability of the drug to trigger cell death through activation of the intrinsic pathway by the release of AIF responsible for mitochondrial permeability and chromatin condensation independent of caspases activation.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis Inducing Factor/metabolism , Apoptosis , Bleomycin/analogs & derivatives , Carcinoma/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Laryngeal Neoplasms/enzymology , Anthracenes/pharmacology , Apoptosis Inducing Factor/drug effects , Bleomycin/pharmacology , Cell Line, Tumor , Colonic Neoplasms/metabolism , Fas Ligand Protein/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Leukemia/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism
14.
Brain Res ; 1285: 30-41, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19555672

ABSTRACT

Activated microglia produce a factor or cocktail of factors that promotes cholinergic neuronal differentiation of undifferentiated precursors in the embryonic basal forebrain (BF) in vitro. To determine whether microglial prostaglandins mediate this action, microglia were stimulated in the presence of the cyclooxygenase inhibitor ibuprofen, and microglial conditioned medium (CM) was used to culture rat BF precursors at embryonic day 15. Choline acetyltransferase (ChAT) activity served as a measure of cholinergic differentiation. While inhibition of prostaglandin biosynthesis did not affect the ability of microglial CM to promote ChAT activity, treatment of microglia with prostaglandin E2 (PGE2) inhibited it. Agonists of E prostanoid receptors EP2 (butaprost) and EP1/3 (sulprostone) mimicked PGE2, while misoprostol (E1-4) actually enhanced the action of CM. PGE2 added directly to BF cultures together with microglial CM also inhibited ChAT activity. While BF cultures expressed all four prostanoid receptors, direct addition of sulprostone but not butaprost mimicked PGE2, suggesting that PGE2 engaged EP1/3 receptors in the BF. Neither PKA inhibition by H89 nor cAMP induction by forskolin or dibutyrl-cAMP altered the action of sulprostone. Sulprostone severely compromised ChAT activity, dendrite number, axonal length and axonal branching, but caspase inhibition did not restore these. However, sulprostone resulted in increased staining intensity and nuclear translocation of apoptosis-inducing factor (AIF) suggesting caspase-independent cell death. We have found that PGE2 action at microglial EP2 receptors inhibits the microglial production of the cholinergic differentiating cocktail, while action at neuronal EP3 receptors has a deleterious effect on cholinergic neurons causing neurite retraction and cell death.


Subject(s)
Apoptosis Inducing Factor/drug effects , Apoptosis/drug effects , Basal Nucleus of Meynert/drug effects , Cholinergic Fibers/drug effects , Prostaglandins/pharmacology , Receptors, Prostaglandin E/agonists , Abortifacient Agents, Nonsteroidal/pharmacology , Animals , Apoptosis/physiology , Apoptosis Inducing Factor/metabolism , Basal Nucleus of Meynert/embryology , Basal Nucleus of Meynert/growth & development , Biomarkers/analysis , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Cholinergic Fibers/metabolism , Cholinergic Fibers/pathology , Dinoprostone/analogs & derivatives , Dinoprostone/metabolism , Dinoprostone/pharmacology , Encephalitis/chemically induced , Encephalitis/metabolism , Encephalitis/physiopathology , Microglia/drug effects , Microglia/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Prostaglandins/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Prostaglandin E/metabolism , Receptors, Prostaglandin E, EP2 Subtype , Receptors, Prostaglandin E, EP3 Subtype
15.
Oncol Rep ; 21(3): 665-71, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212624

ABSTRACT

Capsaicin (trans-8-methyl-N-vanillyl-6-nonenamide), a significant pungent ingredient in a variety of red peppers of the genus Capsicum, is a type of vanilloid. It has been shown to exert biological activities (anticarcinogenic, antimutagenic and chemopreventive) in many cancer cell lines. It was found that capsaicin induces dose-dependent growth inhibition of MCF-7 cells, which does not express caspase-3. In this study, we investigated the molecular mechanism of capsaicin-induced apoptosis in MCF-7 cells. Treatment with capsaicin for 24 h resulted in dose-dependent apoptosis in these cells. After the addition of capsaicin, the levels of reactive oxygen species were reduced slightly in the earlier stage of treatment. Interestingly, an elevation of intracellular calcium ion concentration was detected in the MCF-7 cells. In time course and dosage studies, the mitochondrial membrane potential of MCF-7 cells decreased. However, the change was not significant. It is worth noting that the apoptosis-inducing factor translocated into the cytosol and nucleus from the mitochondria. Our results suggest that capsaicin induces cellular apoptosis through a caspase-independent pathway in MCF-7 cells, and that reactive oxygen species and intracellular calcium ion fluctuation has a minimal role in the process.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Capsaicin/pharmacology , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Blotting, Western , Breast Neoplasms/metabolism , Calcium/metabolism , Caspase 3/metabolism , Cell Line, Tumor , DNA Fragmentation/drug effects , Female , Flow Cytometry , Fluorescent Antibody Technique , Humans , Microscopy, Confocal , Reactive Oxygen Species
16.
Brain Res ; 1247: 28-37, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-18992729

ABSTRACT

EUK-207 is a synthetic superoxide dismutase/catalase mimetic that has been shown to reverse age-related learning deficits and brain oxidative stress in mice. In the present experiments, we tested the effects of EUK-207 on oxygen/glucose deprivation (OGD)-induced cell death in cultured hippocampal slices and on several mechanisms that have been postulated to participate in this process. Cultured hippocampal slices were subjected to 1 h OGD followed by 3 or 24 h recovery in regular medium with glucose and oxygen. Lactate dehydrogenase (LDH) release in culture medium and propidium iodide (PI) uptake in slices were used to evaluate cell viability. When EUK-207 was applied either 1 or 2 h before OGD, OGD-induced LDH release was significantly reduced. When EUK-207 was applied 1 h before OGD and during 24 h recovery, PI uptake was also reduced. OGD-induced accumulation of reactive oxygen species (ROS) was evaluated with the fluorescent probe DCF. DCF fluorescence in slices increased steadily during OGD treatment, rapidly disappeared following return to regular medium before slowly increasing again during the 24 h recovery period. When measured 3 h after OGD, increased ROS levels were significantly reduced by EUK-207. OGD also increased lipid peroxidation levels and this effect was also reduced by EUK-207 6 h following OGD. Cytosolic cytochrome c and nuclear apoptosis-inducing factor (AIF) were increased 3 h after OGD, and the translocation of AIF from mitochondria to nucleus was partly blocked by treatment with EUK-207. In conclusion, EUK-207 provides neuroprotection against OGD-induced cell death in cultured hippocampal slices. As EUK-207 prevents free radical formation and lipid peroxidation, the neuroprotection is related to elimination of free radical generation and lipid peroxidation, as well as to decreased activation of pro-apoptotic factors. Our data support the further clinical evaluation of this class of molecules for the prevention of ischemic cell damage.


Subject(s)
Hypoxia-Ischemia, Brain/drug therapy , Nerve Degeneration/drug therapy , Neurons/drug effects , Organometallic Compounds/pharmacology , Oxidative Stress/drug effects , Superoxide Dismutase/drug effects , Animals , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Catalase/drug effects , Catalase/metabolism , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cytochromes c/metabolism , Fluorescent Dyes , Free Radical Scavengers/pharmacology , Free Radical Scavengers/therapeutic use , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/physiopathology , Hypoxia-Ischemia, Brain/enzymology , Hypoxia-Ischemia, Brain/physiopathology , L-Lactate Dehydrogenase/drug effects , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Nerve Degeneration/enzymology , Nerve Degeneration/etiology , Neurons/enzymology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Organ Culture Techniques , Organometallic Compounds/therapeutic use , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
17.
Clin Chim Acta ; 401(1-2): 76-80, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19061880

ABSTRACT

BACKGROUND: Recent studies have demonstrated that Rho-kinase has been proposed to play an important role in the pathogenesis of heart ischemia/reperfusion (I/R) injury. However, the mechanism of Rho-kinase mediated cardiomyocyte apoptosis in I/R is still not thoroughly understood. METHOD: Studies were performed with female Wistar rats. RESULTS: Ischemia followed by reperfusion caused a significant increase in Rho-kinase, c-Jun NH2-terminal kinase (JNK) and apoptosis-inducing factor (AIF) activity. Administration of fasudil, an inhibitor of Rho-kinase, decreased myocardial infarction size from 59.89+/-3.83% to 38.62+/-2.66% (P<0.05) and cell apoptosis from 32.78+/-5.1% to 17.05+/-4.2% (P<0.05). Western blot analysis showed that administration of fasudil reduced the activation of JNK and attenuated mitochondrial-nuclear translocation of AIF. Additionally, administration of SP600125, an inhibitor of JNK, attenuated mitochondrial-nuclear translocation of AIF. CONCLUSION: The inhibition of Rho-kinase reduced cell apoptosis in I/R in vivo via suppression of JNK-mediated AIF translocation.


Subject(s)
Apoptosis Inducing Factor/metabolism , MAP Kinase Kinase 4/metabolism , Myocardial Infarction/enzymology , Myocytes, Cardiac/enzymology , Reperfusion Injury/enzymology , rho-Associated Kinases/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Anthracenes/pharmacology , Apoptosis , Apoptosis Inducing Factor/drug effects , Female , MAP Kinase Kinase 4/drug effects , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Reperfusion Injury/pathology , rho-Associated Kinases/antagonists & inhibitors
18.
Naunyn Schmiedebergs Arch Pharmacol ; 378(5): 447-57, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18607570

ABSTRACT

Denbinobin is a phenanthraquinone derivative present in the stems of Ephemerantha lonchophylla. We showed that denbinobin induces apoptosis in human colorectal cancer cells (HCT-116) in a concentration-dependent manner. The addition of a pan-caspase inhibitor (zVAD-fmk) did not suppress the denbinobin-induced apoptotic effect, and denbinobin-induced apoptosis was not accompanied by processing of procaspase-3, -6, -7, -9, and -8. However, denbinobin triggered the translocation of the apoptosis-inducing factor (AIF) from the mitochondria into the nucleus. Small interfering RNA targeting of AIF effectively protected HCT-116 cells against denbinobin-induced apoptosis. Denbinobin treatment also caused DNA damage, activation of the p53 tumor suppressor gene, and upregulation of numerous downstream effectors (p21WAF1/CIP1, Bax, PUMA, and NOXA). A HCT-116 xenograft model demonstrated the in vivo efficacy and low toxicity of denbinobin. Taken together, our findings suggest that denbinobin induces apoptosis of human colorectal cancer HCT-116 cells via DNA damage and an AIF-mediated pathway. These results indicate that denbinobin has potential as a novel anticancer agent.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Colorectal Neoplasms/drug therapy , Phenanthrenes/pharmacology , Animals , Anthraquinones/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Colorectal Neoplasms/pathology , DNA Damage/drug effects , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, SCID , Orchidaceae/chemistry , Phenanthrenes/administration & dosage , Xenograft Model Antitumor Assays
19.
Am J Chin Med ; 36(1): 171-84, 2008.
Article in English | MEDLINE | ID: mdl-18306460

ABSTRACT

Natural products including plants, microorganisms and marine life provide rich resources for anticancer drug discovery. The root bark of Hibiscus syriacus has been used as an antipyretic, anthelmintic and antifungal agent in Asia. The antiproliferative effects of H. syriacus on human lung cancer cells were evaluated with bio-assays. The apoptotic activity was detected by Hoechst 33342 DNA staining and annexin V staining. The expression of caspases, p53, apoptosis induced factor (AIF), Bcl-2 and Bax were evaluated with Western blotting. The in vivo anticancer activity was evaluated using A549-xenograft model. The acetone extract of H. syriacus (HS-AE) exhibited a better cytotoxic effect on lung cancer cells than its methanol extract (HS-ME) or water extract (HS-WE). The IC(50) values of HS-AE on A549 (adenocarcinoma), H209 (squamous cell carcinoma) or H661 (large cell carcinoma) lung cancer cells ranged from 14 to 22 microg/ml after 48 hours of treatment. After 48 hours of exposure, HS-AE (15 microg/ml) induced A549 cell apoptosis to 48 +/- 3.6% of the control. Using Western blotting, HS-AE appears to suppress the expression of p53 and AIF. The results of the in vivo study showed that HS-AE suppresses growth in A549 subcutaneous xenograft tumors. These results indicate that HS-AE exerts significant and dose-dependent antiproliferative effects on cancer cells in vitro and in vivo, which prompts us to further evaluate and elucidate the bioactive component(s) of H. syriacus.


Subject(s)
Apoptosis Inducing Factor/genetics , Apoptosis/drug effects , Hibiscus , Lung Neoplasms/drug therapy , Plant Extracts/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis Inducing Factor/drug effects , Cell Cycle/drug effects , Cell Division/drug effects , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Plant Extracts/therapeutic use , Tumor Suppressor Protein p53/drug effects
20.
Rheumatology (Oxford) ; 47(3): 301-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18276737

ABSTRACT

OBJECTIVE: Resveratrol is a naturally occurring polyphenol, which possesses chemotherapeutic potential through its ability to trigger apoptosis. The objective of this study was to investigate the major determinant for the apoptotic cell death induction by resveratrol in fibroblast-like synoviocytes (FLS) derived from patients with RA. METHODS: The effect of resveratrol on apoptotic cell death was quantified in a population of subG1 in RA FLS by flow cytometry. The underlying signalling mechanism for apoptotic death was examined by analysing mitochondrial membrane potential, activation of the caspase cascade and translocation of Bid. RESULTS: We show that activation of caspase-8 is essential for triggering resveratrol-induced apoptotic signalling via the involvement of the mitochondrial pathway in RA FLS. Our findings also suggest that this enhanced apoptosis caused by resveratrol occurred in RA FLS irrespective of p53 status. Exposure to resveratrol caused extensive apoptotic cell death, along with a caspase-dependent (activation of caspase-9 and -3, poly ADPribose polymerase (PARP) cleavage and mitochondrial cytochrome c release) or caspase-independent [translocation of apoptosis-inducing factor (AIF) to the nucleus] signalling pathway. Analysis of upstream signalling events affected by resveratrol revealed that the activated caspase-8 triggered mitochondrial apoptotic events by inducing Bid cleavage without any alteration in the levels of Bax, Bcl-xL or Bcl2. The caspase-8 inhibitor or over-expression of crmA abrogated cell death induced by resveratrol and prevented processing of the downstream cascade. CONCLUSION: The results suggest that resveratrol causes activation of caspase-8, which in turn results in modulation of mitochondrial apoptotic machinery to promote apoptosis of RA FLS.


Subject(s)
Apoptosis Inducing Factor/metabolism , Apoptosis/drug effects , Arthritis, Rheumatoid/physiopathology , Caspase 8/metabolism , Stilbenes/pharmacology , Apoptosis Inducing Factor/drug effects , Arthritis, Rheumatoid/metabolism , Caspase 8/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/drug effects , Fluorescent Antibody Technique , Humans , Membrane Potentials , Mitochondria/physiology , Probability , Resveratrol , Sensitivity and Specificity , Synovial Membrane/cytology , Synovial Membrane/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...