Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Mol Med Rep ; 27(2)2023 Feb.
Article in English | MEDLINE | ID: mdl-36601752

ABSTRACT

The cell­killing potential of most chemotherapeutic agents is enhanced by a temperature elevation. Isofraxidin (IF) is a coumarin compound widely found in plants, such as the Umbelliferae or Chloranthaceae families. IF induces anticancer effects in lung and colorectal cancer. To the best of our knowledge, the combined effects of hyperthermia (HT) and IF on heat­induced apoptosis have not been reported. Acute monocytic leukemia U937 cells were exposed to HT with or without IF pre­treatment. Apoptosis was measured by Annexin V­FITC/PI double staining assay using flow cytometry and cell viability was observed by cell counting kit assay, DNA fragmentation. The mechanism involved in the combination was explored by measuring changes in the mitochondrial membrane potential, (MMP), intracellular ROS generation, expression of apoptosis related protein, and intracellular calcium ion level. It was demonstrated that IF enhanced HT­induced apoptosis in U937 cells. The results demonstrated that combined treatment enhanced mitochondrial membrane potential loss and transient superoxide generation increased protein expression levels of caspase­3, caspase­8 and phosphorylated­JNK and intracellular calcium levels. Moreover, the role of caspases and JNK was confirmed using a pan caspase inhibitor (zVAD­FMK) and JNK inhibitor (SP600125) in U937 cells. Collectively, the data demonstrated that IF enhanced HT­induced apoptosis via a reactive oxygen species mediated mitochondria/caspase­dependent pathway in U937 cells.


Subject(s)
Hyperthermia, Induced , Leukemia, Monocytic, Acute , Humans , U937 Cells , Calcium/metabolism , Apoptosis , Coumarins/pharmacology , Caspases/metabolism , Reactive Oxygen Species/metabolism , Oxidation-Reduction , Membrane Potential, Mitochondrial
2.
Oncol Lett ; 14(1): 1035-1040, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28693270

ABSTRACT

Bufadienolides are constituents of the traditional Chinese medicine Chan Su and are found in toad venom. Cardiovascular side-effects are one of the limiting factors towards developing bufadienolides as chemotherapeutic agents. Thus, in the present study, low doses of bufalin and cinobufotalin, prominent members of the bufadienolides, were investigated for their cytotoxic activity in combination with hyperthermia (HT) or radiation (Rad) therapy. In addition, the underlying mechanism involved was investigated. A DNA fragmentation assay, viability assay and microscopic observation were primarily used to assess the effect of low doses of the two drugs in human lymphoma U937 cells. Furthermore, the effects of these drugs on the mitochondrial membrane potential (MMP) and apoptotic-associated protein activation were investigated. HT/bufadienolide- and RT/bufadienolide-treated samples significantly increased the DNA fragmentation percentile and decreased the MMP, as well as increasing the apoptotic features observed microscopically within a relatively short time (6 h) after treatment. The two combinations affected the expression of important apoptotic markers, including caspase-3 and BH3 interacting domain death agonist. The findings of the current study confirm the additive effect of HT with this group of drugs, directing a novel therapeutic avenue for the clinical use of bufadienolides at lower doses with more restrained cardio toxic side-effects.

3.
Cell Stress Chaperones ; 21(5): 873-81, 2016 09.
Article in English | MEDLINE | ID: mdl-27448221

ABSTRACT

KRAS mutant lung cancers have long been considered as untreatable with drugs. Transforming growth factor-ß-activated kinase 1 (TAK1) appears to play an anti-apoptotic role in response to multiple stresses and has been reported to be a responsive kinase that regulates cell survival in KRAS-dependent cells. In this study, in order to find a useful approach to treat KRAS mutant lung cancer, we focused on the combined effects of 5Z-7-oxozeaenol, a TAK1 inhibitor, with hyperthermia (HT) in KRAS mutant lung cancer cell line A549. Annexin V-FITC/PI assay, cell cycle analysis, and colony formation assay revealed a significant enhancement in apoptosis induced by HT treatment, when the cells were pre-incubated with 5Z-7-oxozeaenol in a dose-dependent manner. The enhanced apoptosis by 5Z-7-oxozeaenol was accompanied by a significant increase in reactive oxygen species (ROS) generation and loss of mitochondrial membrane potential (MMP). In addition, western blot showed that 5Z-7-oxozeaenol enhanced HT-induced expressions of cleaved caspase-3, cleaved caspase-8, and HSP70 and decreased HT-induced expressions of Bcl-2, p-p38, p-JNK, and LC3. Moreover, 5Z-7-oxozeaenol pre-treatment resulted in a marked elevation of intracellular calcium level which might be associated with endoplasmic reticulum (ER) stress-related pathway. Taken together, our data provides further insights of the mechanism of action of 5Z-7-oxozeaenol and HT treatment, and their potential application as a novel approache to treat patients with KRAS mutant lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Lung Neoplasms/therapy , Zearalenone/analogs & derivatives , A549 Cells , Combined Modality Therapy , Drug Screening Assays, Antitumor , Endoplasmic Reticulum Stress , Humans , Hyperthermia, Induced , MAP Kinase Kinase Kinases/antagonists & inhibitors , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/metabolism , Zearalenone/pharmacology
4.
Apoptosis ; 19(11): 1654-63, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25187044

ABSTRACT

Ionizing radiation (IR) can generate reactive oxygen species (ROS). Excessive ROS have the potential to damage cellular macromolecules including DNA, proteins, and lipids and eventually lead to cell death. In this study, we evaluated the potential of arbutin, a drug chosen from a series of traditional herbal medicine by measuring intracellular hydroxyl radical scavenging ability in X-irradiated U937 cells. Arbutin (hydroquinone-ß-D-glucopyranoside), a naturally occurring glucoside of hydroquinone, has been traditionally used to treat pigmentary disorders. However, there are no reports describing the effect of arbutin on IR-induced apoptosis. We confirmed that arbutin can protect cells from apoptosis induced by X-irradiation. The combination of arbutin and X-irradiation could reduce intracellular hydroxyl radical production and prevent mitochondrial membrane potential loss. It also could down-regulate the expression of phospho-JNK, phospho-p38 in whole cell lysate and activate Bax in mitochondria. Arbutin also inhibits cytochrome C release from mitochondria to cytosol. To verify the role of JNK in X-irradiation-induced apoptosis, the cells were pretreated with a JNK inhibitor, and found that JNK inhibitor could reduce apoptosis induced by X-irradiation. Taken together, our data indicate that arbutin plays an anti-apoptotic role via decreasing intracellular hydroxyl radical production, inhibition of Bax-mitochondria pathway and activation of the JNK/p38 MAPK pathway.


Subject(s)
Apoptosis/drug effects , Arbutin/pharmacology , Free Radical Scavengers/pharmacology , Radiation-Protective Agents/pharmacology , Apoptosis/radiation effects , Arbutin/chemistry , Arbutin/metabolism , Caspase 8/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , U937 Cells , fas Receptor/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Apoptosis ; 19(6): 1043-53, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24692054

ABSTRACT

Ionizing radiation (IR) leads to oxidizing events such as excessive reactive oxygen species (ROS) in the exposed cells, resulting in further oxidative damage to lipids, proteins and DNA. To screen the potential radio-protective drug, the intracellular ROS was measured in irradiated U937 cells pretreated with 80 candidate traditional herbal medicine, respectively. Isofraxidin (IF) was one possible radio-protector in these 80 drugs. This study investigated the radio-protective role of IF, a Coumarin compound, in human leukemia cell lines, for the first time. Results indicate that IF protects against IR-induced apoptosis in U937 cells in the time- and concentration- dependent manner. IF decreases IR-induced intracellular ROS generation, especially hydroxyl radicals formation, inhibits IR-induced mitochondrial membrane potential loss and reduces IR-induced high intracellular Ca(2+) levels regardless of ER stress. IF down-regulates the expression of caspase-3, phospho-JNK, phospho-p38 and activates Bax in mitochondria. IF inhibits cytochrome c release from mitochondria to cytosol. IF also moderates IR-induced Fas externalization and caspase-8 activation. IF also exhibits significant protection against IR-induced cell death in other leukemia cell lines such as Molt-4 cells and HL60 cells regardless of p53. Taken together, the data demonstrate that IF protects leukemia cells from radiation-induced apoptosis via ROS/mitochondria pathway in a p53-independent manner.


Subject(s)
Apoptosis/drug effects , Coumarins/pharmacology , Free Radical Scavengers/pharmacology , Mitochondria/metabolism , Radiation-Protective Agents/pharmacology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/radiation effects , Cell Line, Tumor , Coumarins/chemistry , Drug Screening Assays, Antitumor , Free Radical Scavengers/chemistry , Humans , Leukemia , Lymphoma , Membrane Potential, Mitochondrial , Radiation-Protective Agents/chemistry , Signal Transduction , X-Rays
6.
Chem Biol Interact ; 215: 46-53, 2014 May 25.
Article in English | MEDLINE | ID: mdl-24661947

ABSTRACT

To develop a non-toxic enhancer for hyperthermia-induced cell death as a potential cancer treatment, we studied the effect and mechanism of docosahexaenoic acid (DHA) on hyperthermia-induced apoptosis. Treatment with 20µM DHA and 44°C for 10min induced significant apoptosis, increased intracellular reactive oxygen species (ROS), and caspase-3 activation in U937 cells, but heat or DHA alone did not induce notable apoptosis. Decreased mitochondrial transmembrane potentials were dramatically increased by the combined treatment, accompanied by increased pro-apoptotic Bcl-2 family protein tBid, and decreased anti-apoptotic Bcl-2 and Bcl-xL. Combined hyperthermia-DHA treatment induced significant phosphorylation of protein kinase C (PKC)-δ (p-PKC-δ), and apoptosis in a DHA dose-dependent manner. Using both 20µM DHA and 44°C for 10min induced significant PKC-δ cleavage and its translocation to mitochondria. These results were also seen in HeLa cells. However, MAPKs and Akt were not affected by the treatment. In conclusion, DHA enhances hyperthermia-induced apoptosis significantly via a mitochondria-caspase-dependent pathway; its underlying mechanism involves elevated intracellular ROS, mitochondria dysfunction, and PKC-δ activation.


Subject(s)
Apoptosis/drug effects , Docosahexaenoic Acids/pharmacology , Hyperthermia, Induced , Cell Line, Tumor , Docosahexaenoic Acids/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
7.
Eur J Pharmacol ; 723: 99-107, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24321857

ABSTRACT

Hyperthermia is a good therapeutic tool for non-invasive cancer therapy; however, its cytotoxic effects are not sufficient. In the present study, withaferin A (WA), a steroidal lactone derived from the plant Withania somnifera Dunal, has been investigated for its possible enhancing effects on hyperthermia-induced apoptosis. In HeLa cells, treatment with 0.5 or 1.0µM WA at 44°C for 30min induced significant apoptosis accompanied by decreased intracellular GSH/GSSG ratio and caspase-3 activation, while heat or WA alone did not induce such changes. The upregulation in apoptosis was significantly inhibited by glutathione monoethyl ester, a cell permeable glutathione precursor. Mitochondrial transmembrane potentials were dramatically decreased by the combined treatment, with increases in pro-apoptotic Bcl-2-family proteins tBid and Noxa, and downregulation of antiapoptotic Bcl-2 and Mcl-1. Combined treatment with hyperthermia and WA induced significant increases in JNK phosphorylation (p-JNK), and decreases in the phosphorylation of ERK (p-ERK) compared with either treatment alone. These results suggest that WA enhances hyperthermia-induced apoptosis via a mitochondria-caspase-dependent pathway; its underlying mechanism involves elevated intracellular oxidative stress, mitochondria dysfunction, and JNK activation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Hyperthermia, Induced , Withanolides/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Glutathione/metabolism , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/physiology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
8.
Chem Biol Interact ; 199(3): 154-60, 2012 Sep 30.
Article in English | MEDLINE | ID: mdl-22898211

ABSTRACT

Cinobufotalin (CB), one of the bufadienolides prepared from toad venom, was investigated for its cytotoxicity, and the underneath mechanism involved. We primarily utilized DNA fragmentation assay and microscopic observation to assess the effect of various doses of CB in human lymphoma U937 cells. Following that, we investigated other parameters involved in cell death mechanism such as reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and apoptotic proteins activation. HeLa cells were concomitantly used to generalize the data observed. Our results show that CB caused significant DNA fragmentation, decrease of MMP, and an increase in the intracellular Ca(2+) ion and ROS production. In addition, CB induced upregulation of Fas protein, proteolytic activation of cytochrome c, caspase-2, -3, -8 and -9 together with the activation of Bid and Bax. Our findings were further validated using either Fas/FasL antagonist or pan-caspase inhibitor to significantly inhibit CB-induced DNA fragmentation. In our study, we suggest that CB induces caspase dependent cell death in U937 cells, and that Fas plays a role in CB-induced apoptosis. Altogether, our data provides novel insights of the mechanism of action of CB and its potential as a future chemotherapeutic agent.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bufanolides/pharmacology , Amphibian Venoms/chemistry , Animals , Antineoplastic Agents/isolation & purification , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/metabolism , Bufanolides/isolation & purification , Calcium/metabolism , Caspases/metabolism , DNA Fragmentation/drug effects , Drug Screening Assays, Antitumor , HeLa Cells , Humans , Medicine, Chinese Traditional , Membrane Potential, Mitochondrial/drug effects , Metabolic Networks and Pathways , Reactive Oxygen Species/metabolism , U937 Cells , bcl-2-Associated X Protein/metabolism , fas Receptor/metabolism
9.
Int J Hyperthermia ; 25(5): 364-73, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19551546

ABSTRACT

Sanazole has been tested clinically as a hypoxic cell radiosensitizer. The aim of the present study was to investigate whether sanazole enhances apoptosis induced by hyperthermia at 44 degrees C for 20 min in human lymphoma U937 cells. Sanazole alone induced continuous increase in the intracellular superoxide generation in a time-dependent manner and transient increase in the peroxide formation, which further were enhanced at 1 hour after HT treatment. Moreover, when the cells were treated first with 10 mM sanazole for 40 min, exposed to HT at 44 degrees C for 20 min and the cells were further treated with the drug at 37 degrees C for 6 h, a significant enhancement of HT-induced apoptosis was evidenced by DNA fragmentation, morphological changes and phosphatidylserine externalization. Studying the apoptotic pathways involved in this enhancement, we found that loss of the mitochondrial membrane potential, release of cytochrome c from mitochondria to cytosol, and activation of caspase-3 and caspase-8 was enhanced significantly in the U937 cells after the combined treatment. Moreover, this combination enhanced activation of Bid, and down regulation of Hsp70. In addition, an increase in the intracellular Ca(2+) concentration ([Ca(2+)](i)), and externalization of Fas were observed immediately after sanazole and HT treatment. Our data indicate that sanazole can enhance the hyperthermia induced-apoptosis through the Fas-caspase-8- and [Ca(2+)](i)-dependent apoptotic pathways. In addition, the down regulation of Hsp70 contributed to this enhancement.


Subject(s)
Apoptosis/drug effects , Hyperthermia, Induced , Triazoles/pharmacology , Apoptosis/physiology , Calcium Signaling/drug effects , Caspases/drug effects , Caspases/metabolism , Humans , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , U937 Cells
10.
Anticancer Res ; 28(3A): 1693-700, 2008.
Article in English | MEDLINE | ID: mdl-18630528

ABSTRACT

AIM: The possible enhancing effect of the combined use of sodium butyrate (SB) and hyperthermia to kill HCT 116 cells was evaluated. MATERIALS AND METHODS: HCT 116 cells were subjected to SB (1 mM) treatment followed by hyperthermia (44 degrees C 60 min) and the effects on cell death, cell proliferation and the cell cycle were examined. Apoptosis-indicating protein expressions and intracellular superoxide formation were also analysed. RESULTS: A marked reduction in the growth rate of the combined-treatment group was observed compared to those of the single-treatment groups. This involved the increased expression of p53 and p21, the alteration of the balance of anti- and proapoptotic Bcl-2 family proteins and enhanced superoxide formation. However, the death receptor pathway played no role. CONCLUSION: Hyperthermia synergistically promoted cell death induced by SB. Thus, the combined treatment led to mutual potentiation of the killing effects of each agent.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Colorectal Neoplasms/therapy , Hyperthermia, Induced/methods , BH3 Interacting Domain Death Agonist Protein/biosynthesis , Caspase 3/metabolism , Caspase 8/metabolism , Cell Cycle/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Combined Modality Therapy , DNA Fragmentation , HCT116 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Superoxides/metabolism , bcl-2 Homologous Antagonist-Killer Protein/biosynthesis , bcl-X Protein/biosynthesis
11.
Apoptosis ; 13(3): 448-61, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18224486

ABSTRACT

The aim of this study was to examine whether, a new synthesized class of benzocycloalkene derivatives (BCs), enhances apoptosis induced by hyperthermia. The combined effects of hyperthermia (44 degrees C, 20 min) and BCs on apoptosis in human lymphoma U937 cells were investigated. Among the tested compounds (BC1 approximately 9), the combined treatment of 10 muM BC2 or BC4 and hyperthermia showed the largest potency to induce DNA fragmentation at 6 h after hyperthermia. And enhancement of hyperthermia-induced apoptosis by BC2 or BC4 in a dose-dependent manner was observed. When the cells were treated first with BC2 or BC4 at a nontoxic concentration of 20 muM, and exposed to hyperthermia afterwards, a significant enhancement of heat-induced apoptosis was evidenced by DNA fragmentation, morphological changes and phosphatidylserine externalization. Flow cytometry revealed an increase of intracellular superoxide due to BC2 or BC4, which was further increased when hyperthermia was combined. Mitochondrial membrane potential was decreased and the activation of caspase-3 and caspase-8 was enhanced in the cells treated with the combination. The activation of Bid, but no change of Bax and Bcl-2 were observed after the combined treatment. The release of cytochrome c from mitochondria to cytosol, which was induced by hyperthermia, was enhanced by BC2 or BC4. An increase in the intracellular Ca2+ concentration [Ca2+](i), externalization of Fas, and decrease in Hsp70 were observed following the combined treatment. These results indicate that the intracellular superoxide generated by BC2 or BC4 is involved in the enhancement of apoptosis through Fas-mitochondria caspase and [Ca2+](i)-dependent pathways, and a decrease in Hsp70 also contributed to the enhancement of apoptosis.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Hyperthermia, Induced/adverse effects , Polycyclic Aromatic Hydrocarbons/pharmacology , Calcium/physiology , Caspases/metabolism , DNA Fragmentation/drug effects , HSP70 Heat-Shock Proteins/biosynthesis , Humans , Membrane Potential, Mitochondrial/drug effects , Oxidative Stress/drug effects , Polycyclic Compounds/pharmacology , Reactive Oxygen Species/metabolism , U937 Cells , fas Receptor/metabolism
12.
Int J Hyperthermia ; 23(6): 529-37, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17952766

ABSTRACT

PURPOSE AND BACKGROUND: Paeoniflorin (PF) isolated from peony root (Paeoniae radix) has been used as a herbal medicine in East Asia for its anti-allergic, anti-inflammatory, and immunoregulatory effects. PF is known to cause apoptosis and to be a chemical heat shock protein (HSP) inducer. With this information, the effects on the gene expression in human leukemia U937 cells treated with PF were investigated. METHODS: U937 cells, a human myelomonocytic cell line, were treated with PF at different concentrations (0-640 microg/ml). Expression level of Hsp70 was monitored by Western blotting. Gene expression was evaluated using high-density oligonucleotide microarrays and computational gene expression analysis tools and the results were verified by real-time quantitative PCR. RESULTS: Although cell viability was not affected after PF treatment at a high concentration of 640 microg/ml, PF treatment (80-640 microg/ml) significantly elevated Hsp70 expression in a concentration-dependent manner. When the cells were treated with PF (160 microg/ml; 30 min), 35 up-regulated and 29 down-regulated genes were identified. Among the differentially expressed genes, a significant genetic network containing CDC2, FOSL1 and EGR1 was associated with biological functions such as cell death, gene expression or cellular growth and proliferation. CONCLUSION: The present results indicate that PF affects the expression of many genes including Hsp70 and will provide a better understanding on the molecular mechanism of action of this compound in inducing HSPs in cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Benzoates/pharmacology , Bridged-Ring Compounds/pharmacology , Gene Expression Regulation/drug effects , Glucosides/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Apoptosis/drug effects , Gene Expression Profiling , HSP70 Heat-Shock Proteins/drug effects , HSP70 Heat-Shock Proteins/genetics , Humans , Monoterpenes , Oligonucleotide Array Sequence Analysis , U937 Cells , Up-Regulation
13.
Apoptosis ; 12(8): 1523-32, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17458712

ABSTRACT

The combined effects of hyperthermia (44 degrees C, 20 min) or X-rays (10 Gy) and a new class of furan-fused tetracyclic synthesized compounds (DFs), on apoptosis in human lymphoma U937 cells were investigated. Among the tested compounds (DF1 approximately 6), the combined treatment of 10 microM DF with TIPS (triisopropylsilyloxy) (Designated #3 DF3) and hyperthermia showed the largest potency to induce DNA fragmentation at 6 h after hyperthermia but no enhancement was observed if it was combined with X-rays. Enhancement of hyperthermia-induced apoptosis by DF3 in a dose-dependent manner was observed. When the cells were treated first with DF3 at a nontoxic concentration of 20 microM, and exposed to hyperthermia afterwards, a significant enhancement of heat-induced apoptosis was evidenced by DNA fragmentation, morphological changes and phosphatidylserine externalization. The activation of Bid, but no change of Bax and Bcl-2 were observed after the combined treatment. The release of cytochrome c from mitochondria to cytosol, which was induced by hyperthermia, was enhanced by DF3. Mitochondrial transmembrane potential was decreased and the activation of caspase-3 and caspase-8 was enhanced in the cells treated with the combination. Externalization of Fas was observed following the combined treatment. Flow cytometry revealed rapid and sustained increase of intracellular superoxide due to DF3, and showed subsequent and transient increase in the formation of intracellular hydrogen peroxide (H(2)O(2)), which was further increased when hyperthermia was combined. These results indicate that the intracellular superoxide and H(2)O(2) generated by DF3 enhance the hyperthermia-induced apoptosis via the Fas-mediated mitochondrial caspase-dependent pathway.


Subject(s)
Apoptosis/drug effects , Furans/therapeutic use , Hyperthermia, Induced , Lymphoma, Large B-Cell, Diffuse/therapy , Apoptosis Regulatory Proteins/metabolism , Caspases/metabolism , Combined Modality Therapy , Furans/chemical synthesis , Furans/classification , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/therapeutic use , Humans , Lymphoma, Large B-Cell, Diffuse/pathology , Mitochondria/enzymology , Models, Biological , Reactive Oxygen Species/metabolism , Signal Transduction , U937 Cells , X-Ray Therapy , fas Receptor/metabolism
14.
J Biol Chem ; 277(21): 18986-93, 2002 May 24.
Article in English | MEDLINE | ID: mdl-11861640

ABSTRACT

The combined effects of hyperthermia at 44 degrees C and local anesthetics on apoptosis in human histiocytic lymphoma U937 cells were investigated. When the cells were exposed to hyperthermia for l0 min marginal DNA fragmentation and nuclear fragmentation were observed. In the presence of amide-type local anesthetics further enhancement was found depending on concentration. The order of the concentration required for maximum induction was the reverse order of the lipophilicity (prilocaine > lidocaine > bupivacaine). Western blotting revealed that in hyperthermia there was initial release of Ca(2+) from the intracellular store site as indicated by increased expression of the type 1 inositol-1,4,5-trisphosphate receptor. However, the combination with lidocaine did not induce any further enhancement. Lidocaine enhanced the decrease in ATP content and the increase in intracellular Ca(2+) concentration in individual cells induced by hyperthermia. In addition, superoxide formation, decrease in the mitochondrial membrane potential, and activation of intracellular caspase-3 were found in the cells treated with hyperthermia and lidocaine. All of these were suppressed in part in the presence of the intracellular Ca(2+) ion chelator BAPTA-AM (bis-(O-aminophenoxy)-ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl). The present results indicate that local anesthetics at optimal concentrations enhance hyperthermia-induced apoptosis via Ca(2+)- and mitochondria-dependent pathways. Initial release of Ca(2+) from intracellular store sites caused by hyperthermia and followed by the subsequent increase in the intracellular Ca(2+) concentration and the additional activation of the mitochondrial caspase-dependent pathway (partly regulated by intracellular Ca(2+) concentration) plays a crucial role in the enhancement of apoptosis induced by the combination of hyperthermia and lidocaine.


Subject(s)
Anesthetics, Local/pharmacology , Apoptosis , Egtazic Acid/analogs & derivatives , Hyperthermia, Induced , Lymphoma, Large B-Cell, Diffuse/pathology , Apoptosis/drug effects , Blotting, Western , Calcium/metabolism , Caspases/metabolism , Egtazic Acid/pharmacology , Flow Cytometry , Humans , Lymphoma, Large B-Cell, Diffuse/metabolism , Mitochondria/enzymology , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL