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










Publication year range
1.
Cancer Lett ; 352(2): 214-9, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24997432

ABSTRACT

The present study evaluated the cytotoxic activity of methyl jasmonate (MJ) in endometrial cancer cells and examined the hypothesis that the apoptotic and anti-proliferative actions of MJ in these cell lines can be enhanced by co-targeting the insulin-like growth factor-1 receptor (IGF1R) signaling pathway. MJ had a potent pro-apoptotic effect and exhibited significant toxicity in all cell lines tested. MJ in combination with NVP-AEW541, a selective IGF1R tyrosine kinase inhibitor, had significantly increased cytotoxicity. MJ decreased IGF1R phosphorylation, however, it enhanced AKT phosphorylation and abolished the anti-apoptotic effect of IGF1. These findings suggest that combined IGF1R inhibitor and MJ administration may constitute an attractive modality for treating endometrial cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Endometrial Neoplasms/enzymology , Receptor, IGF Type 1/antagonists & inhibitors , Acetates/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclopentanes/pharmacology , Dose-Response Relationship, Drug , Endometrial Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Oxylipins/pharmacology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, IGF Type 1/metabolism , Time Factors
2.
Curr Pharm Biotechnol ; 14(3): 331-41, 2013.
Article in English | MEDLINE | ID: mdl-22201595

ABSTRACT

Mitochondria are known to play a key role in various cellular processes essential to both the life and death of cells, including calcium homeostasis, programmed cell death, and energy metabolism. Over 80 years ago, Otto Warburg discovered that in contrast to normal cells which produce most of their ATP via mitochondrial oxidative phosphorylation, cancer cells preferentially utilize glycolysis for production of ATP, a phenomenon known today as the "Warburg effect", and one which has been of great importance in the emergence of novel drugs and chemotherapeutic agents specifically targeting cancer cells. Several groups have reported in recent years that members of the plant stress hormones family of jasmonates, and some of their synthetic derivatives, exhibit anti-cancer activity in vitro and in vivo. Jasmonates have been shown to act directly on mitochondria of cancer cells, leading to mitochondrial swelling, membrane depolarization and cytochrome c release. Throughout the last few years, different groups have demonstrated that combination of jasmonates and various cytotoxic and chemotherapeutic agents yielded a synergistic cytotoxic effect. These results have been demonstrated in a variety of different cancer cell lines and may provide a strong basis for future clinical treatments which involve combination of MJ and different anti-cancerous agents. The potential synergistic effect may allow reduction of the administered dose, decrease of unwanted side effects, and reduction of the likelihood that the tumor will display resistance to the combined therapy.


Subject(s)
Acetates/administration & dosage , Antineoplastic Agents/administration & dosage , Cyclopentanes/administration & dosage , Neoplasms/drug therapy , Oxylipins/administration & dosage , Animals , Drug Combinations , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/metabolism
3.
J Mol Med (Berl) ; 91(3): 357-68, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23052480

ABSTRACT

Ovarian carcinoma patients are initially responsive to platinum-based therapy, but eventually become refractory to treatment due to the development of platinum chemoresistance. Elevated levels of interleukin-6 (IL-6) in the sera and ascites of these patients predict poor clinical outcome. Our goal was to analyze the interaction between cisplatin and cisplatin-resistant ovarian cancer cells, and to identify means of circumventing platinum resistance. We studied ovarian carcinoma cell lines and cells drawn from ovarian carcinoma patients. Gene array analyses were performed on ovarian carcinoma cells upon treatment with cisplatin, and the results were validated by ELISA and Western blotting (WB). Cytotoxicity assays were performed on anti-IL-6 Ab-, IL-6-, and cellular inhibitor of apoptosis 2 (cIAP-2) siRNA-treated cells, following cisplatin addition. Our results revealed a highly significant increase in IL-6 and cIAP-2 mRNA and protein levels upon treatment with cisplatin. WB analysis of cisplatin-treated cells exhibited decreased cIAP-2 expression level following anti-IL-6 Ab addition. Furthermore, IL-6 by itself, significantly increased cIAP-2 levels in ovarian carcinoma cells. Finally, cytotoxicity assays showed sensitization to cisplatin following the addition of IL-6 and cIAP-2 inhibitors. In conclusion, cisplatin treatment of ovarian carcinoma cells upregulates IL-6 and cIAP-2 levels while their inhibition significantly sensitizes them to cisplatin. Here, we present cIAP-2 as a novel inducer of platinum resistance in ovarian carcinoma cells, and suggest an axis beginning with an encounter between cisplatin and these cells, mediated sequentially by IL-6 and cIAP-2, resulting in cisplatin resistance. Consequently, we propose that combining IL-6/cIAP-2 inhibitors with cisplatin will provide new hope for ovarian carcinoma patients by improving the current treatment.


Subject(s)
Cisplatin/administration & dosage , Drug Resistance, Neoplasm/genetics , Inhibitor of Apoptosis Proteins/genetics , Interleukin-6/genetics , Ovarian Neoplasms/drug therapy , Apoptosis/drug effects , Baculoviral IAP Repeat-Containing 3 Protein , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Small Interfering , Ubiquitin-Protein Ligases
4.
Invest New Drugs ; 31(2): 333-44, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22956285

ABSTRACT

Current therapies for treatment of advanced cervical cancer involve the use of cisplatin, often in combination with radiotherapy. These treatments do not lead to a high survival rate and furthermore, serious side effects are dose-limiting factors. Methyl jasmonate (MJ) was recently identified as potent and selective cytotoxic agent towards cervical cancer cells. In the present study we evaluated the effectiveness of combined treatments of MJ with cisplatin or X-irradiation on a variety of cervical cancer cells including SiHa, CaSki, HeLa and C33A. Cytotoxicity of alpha particles, emitted from (224)Ra atoms, was also evaluated as a single agent and in combination with MJ. Cooperation between MJ and cisplatin in reducing cell viability (XTT assays) and survival (clonogenicity assays) was exhibited towards several cancer cell lines at a range of combination doses. MJ effectively cooperated also with X-ray irradiation, significantly lowering the radiation doses required to inhibit cell survival (ID50) of all tested cells lines. We show for the first time, that alpha irradiation selectively reduced cell viability and survival of cervical cancer cells. Lower doses of α irradiation were required as compared to X-irradiation to inhibit cell survival. Cooperation with MJ was demonstrated in part of the cancer cell lines. In conclusion, our studies point to α irradiation and MJ, novel anticancer agents, as potent candidates for treatment of cervical cancer, in single agent regiments and in combination. MJ can be added also to conventional X-ray and cisplatin therapies to increase their cytotoxic effect while lowering the effective dose.


Subject(s)
Acetates/pharmacology , Alpha Particles , Apoptosis/drug effects , Apoptosis/radiation effects , Cisplatin/pharmacology , Cyclopentanes/pharmacology , Gamma Rays , Oxylipins/pharmacology , Uterine Cervical Neoplasms/pathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Female , Humans , Plant Growth Regulators/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tumor Cells, Cultured , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/radiotherapy
5.
Cancer Lett ; 319(1): 31-8, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22198483

ABSTRACT

The present study further investigated the mode of action of methyl jasmonate (MJ) in different cervical cancer cell lines. We show that in addition to the short term cytotoxicity, MJ effectively reduced the survival of cervical cancer cells (clonogenicity assays). MJ induced apoptosis in all cervical cancer cells. In some cell lines, MJ caused elevation of the mitochondrial superoxide anion, notably, in HeLa and CaSki. Changes in the expression of p53 and bax were variable, yet, downregulation of survivin was common to all cervical cancer cells. MJ significantly reduced the levels of the human papillomavirus (HPV) E6 and E7 proteins without alteration of the mRNA levels. Moreover, ectopic expression of E6, E7 or both in cervical cancer cells that lack HPV (C33A), did not alter significantly their response to MJ. Our studies point to MJ as an effective anticancer agent against a variety of cervical cancer cells acting through shared and different pathways to induce cell death regardless of the presence of HPV.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cyclopentanes/pharmacology , DNA-Binding Proteins/drug effects , Down-Regulation , Inhibitor of Apoptosis Proteins/drug effects , Oncogene Proteins, Viral/drug effects , Oxylipins/pharmacology , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/drug effects , Uterine Cervical Neoplasms/drug therapy , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Drug Screening Assays, Antitumor , Female , Humans , Inhibitor of Apoptosis Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Repressor Proteins/metabolism , Survivin , Uterine Cervical Neoplasms/metabolism
6.
ACS Med Chem Lett ; 2(7): 528-31, 2011 Jul 14.
Article in English | MEDLINE | ID: mdl-24900344

ABSTRACT

The cytotoxic activity of aloe-emodin (AE), a natural anthranoid that readily permeates anthracycline-resistant tumor cells, was improved by the attachment of an amino-sugar unit to its anthraquinone core. The new class of AE glycosides (AEGs) showed a significant improvement in cytotoxicity-up to more than 2 orders of magnitude greater than those of AE and the clinically used anthracycline doxorubicin (DOX)-against several cancer cell lines with different levels of DOX resistance. Incubation with the synthetic AEGs induced cell death in less than one cell cycle, indicating that these compounds do not directly target the cell division mechanism. Confocal microscopy provided evidence that unlike DOX, AEGs accumulated in anthracycline-resistant tumor cells in which resistance is conferred by P-glycoprotein efflux pumps. The results of this study demonstrate that AEGs may serve as a promising scaffold for the development of cytotoxic agents capable of overcoming anthracycline resistance in tumor cells.

7.
Malar J ; 9: 5, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20051114

ABSTRACT

BACKGROUND: Plasmodium and Schistosoma are two of the most common parasites in tropical areas. Deregulation of the immune response to Plasmodium falciparum, characterized by a Th1 response, leads to cerebral malaria (CM), while a Th2 response accompanies chronic schistosomiasis. METHODS: The development of CM was examined in mice with concomitant Schistosoma mansoni and Plasmodium berghei ANKA infections. The effect of S. mansoni egg antigen injection on disease development and survival was also determined. Cytokine serum levels were estimated using ELISA. Statistical analysis was performed using t-test. RESULTS: The results demonstrate that concomitant S. mansoni and P. berghei ANKA infection leads to a reduction in CM. This effect is dependent on infection schedule and infecting cercariae number, and is correlated with a Th2 response. Schistosomal egg antigen injection delays the death of Plasmodium-infected mice, indicating immune involvement. CONCLUSIONS: This research supports previous claims of a protective effect of helminth infection on CM development. The presence of multiple parasitic infections in patients from endemic areas should therefore be carefully noted in clinical trials, and in the development of standard treatment protocols for malaria. Defined helminth antigens may be considered for alleviation of immunopathological symptoms.


Subject(s)
Malaria, Cerebral/complications , Malaria, Cerebral/epidemiology , Plasmodium falciparum/pathogenicity , Schistosomiasis mansoni/complications , Animals , Cytokines/blood , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Female , Humans , Incidence , Malaria, Cerebral/parasitology , Malaria, Cerebral/pathology , Male , Mice , Plasmodium berghei/immunology , Plasmodium berghei/pathogenicity , Plasmodium falciparum/immunology , Survival Analysis
8.
Phytochemistry ; 70(13-14): 1600-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19660769

ABSTRACT

Jasmonates act as signal transduction intermediates when plants are subjected to environmental stresses such as UV radiation, osmotic shock and heat. In the past few years several groups have reported that jasmonates exhibit anti-cancer activity in vitro and in vivo and induce growth inhibition in cancer cells, while leaving the non-transformed cells intact. Recently, jasmonates were also discovered to have cytotoxic effects towards metastatic melanoma both in vitro and in vivo. Three mechanisms of action have been proposed to explain this anti-cancer activity. The bio-energetic mechanism - jasmonates induce severe ATP depletion in cancer cells via mitochondrial perturbation. Furthermore, methyl jasmonate (MJ) has the ability to detach hexokinase from the mitochondria. Second, jasmonates induce re-differentiation in human myeloid leukemia cells via mitogen-activated protein kinase (MAPK) activity and were found to act similar to the cytokinin isopentenyladenine (IPA). Third, jasmonates induce apoptosis in lung carcinoma cells via the generation of hydrogen peroxide, and pro-apoptotic proteins of the Bcl-2 family. Combination of MJ with the glycolysis inhibitor 2-deoxy-d-glucose (2DG) and with four conventional chemotherapeutic drugs resulted in super-additive cytotoxic effects on several types of cancer cells. Finally, jasmonates have the ability to induce death in spite of drug-resistance conferred by either p53 mutation or P-glycoprotein (P-gp) over-expression. In summary, the jasmonates are anti-cancer agents that exhibit selective cytotoxicity towards cancer cells, and thus present hope for the development of cancer therapeutics.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cyclopentanes/pharmacology , Oxylipins/pharmacology , Acetates/metabolism , Acetates/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclopentanes/metabolism , Cyclopentanes/therapeutic use , Humans , Models, Biological , Neoplasms/drug therapy , Oxylipins/metabolism , Oxylipins/therapeutic use , Plant Growth Regulators/pharmacology
9.
Neoplasia ; 10(11): 1303-13, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18953440

ABSTRACT

Methyl jasmonate (MJ) acts both in vitro and in vivo against various cancer cell lines. Activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway results in decreased susceptibility to cytotoxic agents in many types of cancer cells. We found a strong inverse correlation between the basal level of phospho-Akt (pAkt) and the sensitivity to MJ among sarcoma cell lines. Nevertheless, levels of pAkt increased in two sarcoma cell lines, MCA-105 and SaOS-2, after MJ treatment. Treatment of both cell lines with PI3K/Akt pathway inhibitors in combination with MJ resulted in a synergistic cytotoxic effect. Moreover, cells transfected with a constitutively active Akt were less susceptible to MJ-induced cytotoxicity in comparison with cells transfected with an inactive form of Akt. Taken together, these data suggest that the increase in pAkt after treatment with MJ played a protective role. Because it has been shown that the antiapoptotic effects of Akt are dependent on glycolysis, we examined the role of glucose metabolism in activation of Akt and the subsequent resistance of the cell lines to MJ. 2-Deoxy-d-glucose, a glycolysis inhibitor, decreased the levels of pAkt and was able to attenuate the MJ-induced elevation in pAkt. Accordingly, the presence of glucose attenuated MJ-induced cytotoxicity. Moreover, treatment with 2-deoxy-d-glucose in combination with MJ resulted in a synergistic cytotoxic effect. In conclusion, the PI3K/Akt pathway plays a critical role in the resistance of MCA-105 and SaOS-2 sarcoma cell lines toward MJ-induced cytotoxicity.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Cyclopentanes/pharmacology , Drug Resistance, Neoplasm , Oxylipins/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sarcoma/metabolism , Sarcoma/pathology , Animals , Blotting, Western , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Deoxyglucose/pharmacology , Flow Cytometry , Glycolysis , Humans , Mice , Proto-Oncogene Proteins c-akt/genetics
10.
Cancer Lett ; 271(1): 34-46, 2008 Nov 18.
Article in English | MEDLINE | ID: mdl-18599197

ABSTRACT

In the present study the effectiveness of methyl jasmonate (MJ) against cervical cancer cell lines was investigated. We show that MJ is cytotoxic to a range of cervical cancer lines including SiHa, CaSki and HeLa that carry human papillomavirus (HPV) DNA and wild type p53, and C33A that is negative for HPV and contains mutant p53. Primary human foreskin keratinocytes were almost resistant to the drug. Cytotoxicity of MJ was dose and time dependent, and associated mainly with the induction of cell death and to a less extent with inhibition of cell growth. Cell death induced by MJ displayed features characteristic to both apoptosis and necrosis, and was associated with different changes in the levels of p53, p21, bcl-2 and bax in the various cervical cancer lines. In conclusion, MJ a novel anticancer agent, acts via multiple pathways to induce death of cervical cancer cells, thus making it a promising candidate for treatment of cervical cancer.


Subject(s)
Acetates/pharmacology , Cell Death/drug effects , Cyclopentanes/pharmacology , Oxylipins/pharmacology , Uterine Cervical Neoplasms/pathology , Adenosine Triphosphate/metabolism , Blotting, Western , Cell Division/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , Neoplasm Proteins/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
11.
Int J Parasitol ; 38(8-9): 959-68, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18294640

ABSTRACT

Jasmonates are a group of small lipids produced in plants and function as stress hormones. They are selectively cytotoxic against cancer cells. Methyl jasmonate (MJ), one of the naturally occurring jasmonates, has direct mitochondriotoxic effects, strongly suggesting that mitochondria are target organelles of jasmonates. We have previously shown that jasmonates are cytotoxic to two human parasites -Schistosoma mansoni and Plasmodium falciparum. Both the cancer cells and the parasites mentioned above possess mitochondria. The present work aimed to examine whether jasmonates are able to damage cells lacking mitochondria, e.g., some unicellular human parasitic flagellates. We found that MJ induced death of the amitochondriate Trichomonas vaginalis parasites. MJ caused fragmentation and condensation of the DNA of T. vaginalis, resembling phenomena associated with apoptotic death. However, DNA laddering, a sub-G(1) cell cycle stage peak and caspase-3 activation were not observed. Thus, MJ-induced T. vaginalis cell death appears to be non-apoptotic. We found that MJ induced cell cycle block at the G(2)/M phase in T. vaginalis, similar to the effect of metronidazole. We examined the influence of MJ on the bioenergetic pathways of T. vaginalis, and found that depletion of ATP did not precede death of the parasites, but rather reflected it. Nevertheless, 2-deoxy-d-glucose, a glycolysis blocker, was synergistic with MJ in causing death of T. vaginalis cells, suggesting that MJ does perturb the bioenergetic homeostasis of the parasites. Finally, MJ was found to be cytotoxic towards a metronidazole-resistant strain of T. vaginalis (ATCC 50143), suggesting that it may be effective for the treatment of nitroimidazole-refractory trichomoniasis. In conclusion, MJ was found to exhibit mitochondria-independent cytotoxicity and presents a potentially novel agent against T. vaginalis.


Subject(s)
Acetates/pharmacology , Apoptosis/physiology , Cell Cycle/drug effects , Cyclopentanes/pharmacology , Oxylipins/pharmacology , Plant Growth Regulators/pharmacology , Trichomonas vaginalis/drug effects , Animals , Antiprotozoal Agents/pharmacology , Apoptosis/drug effects , Humans , Metronidazole/pharmacology , Mice , Mitochondria , Trichomonas vaginalis/growth & development
12.
J Bioenerg Biomembr ; 39(1): 51-7, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17549642

ABSTRACT

Jasmonates are plant stress hormones that induce suppression of proliferation and death in cancer cells, while being selectively inactive towards non-transformed cells. Jasmonates can overcome apoptotic blocks and exert cytotoxic effects on drug-resistant cells expressing p53 mutations. Jasmonates induce a rapid depletion of ATP in cancer cells. Indeed, this steep drop occurs when no signs of cell death are detectable yet. Experiments using modulators of ATP synthesis via glycolysis or oxidative phosphorylation suggest that the latter is the pathway suppressed by jasmonates. Consequently, the direct effects of jasmonates on mitochondria were evaluated. Jasmonates induced cytochrome c release and swelling in mitochondria isolated from cancer cells but not from normal ones. Thus, the selectivity of jasmonates against cancer cells is rooted at the mitochondrial level, and probably exploits differences between mitochondria from normal versus cancer cells. These findings position jasmonates as promising anti-cancer drugs acting via energetic depletion in neoplastic cells.


Subject(s)
Adenosine Triphosphate/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cyclopentanes/pharmacology , Mitochondria/physiology , Neoplasms/drug therapy , Oxidative Phosphorylation/drug effects , Antineoplastic Agents, Phytogenic/therapeutic use , Cyclopentanes/therapeutic use , Energy Metabolism , Humans , Mitochondria/drug effects , Oxylipins , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Cancer Lett ; 245(1-2): 1-10, 2007 Jan 08.
Article in English | MEDLINE | ID: mdl-16600475

ABSTRACT

Several groups have reported in recent years that members of the plant stress hormones family of jasmonates, and some of their synthetic derivatives, exhibit anti-cancer activity in vitro and in vivo. Jasmonates increased the life span of EL-4 lymphoma-bearing mice, and exhibited selective cytotoxicity towards cancer cells while sparing normal blood lymphocytes, even when the latter were part of a mixed population of leukemic and normal cells drawn from the blood of chronic lymphocytic leukemia (CLL) patients. Jasmonates join a growing number of old and new cancer chemotherapeutic compounds of plant origin. Three mechanisms of action have been proposed to explain the anti-cancer activity of jasmonates. These include: (1) The bio-energetic mechanism-jasmonates induce severe ATP depletion in cancer cells via mitochondrial perturbation; (2) The re-differentiation mechanism-jasmonates induce re-differentiation in human myeloid leukemia cells via mitogen-activated protein kinase (MAPK) activity; (3) The reactive oxygen species (ROS)-mediated mechanism-jasmonates induce apoptosis in lung carcinoma cells via the generation of hydrogen peroxide, and pro-apoptotic proteins of the Bcl-2 family. Several similarities between the effects of jasmonates on plant and cancer cells have been recorded, suggesting that additional analysis of jasmonate effects in plant cells may contribute to a deeper understanding of the anti-cancer actions of these compounds. Those similarities include: induction of cell death, suppression of proliferation and cell cycle arrest, MAPK induction, ROS generation, and enhancement of heat-shock proteins (HSP) expression. Finally, jasmonates can induce death in drug-resistant cells. The drug resistance was conferred by either p53 mutation or P-glycoprotein (P-gp) over-expression. In summary, the jasmonate family of novel anti-cancer agents presents new hope for the development of cancer therapeutics, which should attract further scientific and pharmaceutical interest.


Subject(s)
Cyclopentanes/therapeutic use , Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cyclopentanes/chemistry , Cyclopentanes/pharmacology , Humans , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Neoplasms/metabolism , Neoplasms/pathology , Oxylipins , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
14.
Cancer Lett ; 250(2): 300-10, 2007 Jun 08.
Article in English | MEDLINE | ID: mdl-17125916

ABSTRACT

The anti-cancer agent methyl jasmonate (MJ) acts in vitro and in vivo against various cancer cell lines, as well as leukemic cells from chronic lymphocytic leukemia (CLL) patients. Given the importance of multi-agent combinations in cancer chemotherapy, the purpose of this study was to identify super-additive combinations of MJ and currently-available chemotherapeutic drugs. We identified such cooperative effects in six cell lines arising from different major types of malignancies, i.e., breast, lung, prostate and pancreas carcinomas as well as leukemia. The chemotherapeutic drugs tested were adriamycin, taxol, BCNU and cisplatin. For instance, MJ exhibited strong cooperative effects with BCNU in MIA PaCa-2 pancreatic carcinoma cells. Furthermore, MJ enhanced significantly (pV=0.028) the anti-leukemic effect of adriamycin in vivo, in a CLL mouse model. Finally, MJ cooperated with the glycolysis inhibitor 2-deoxy-D-glucose in inducing death of several types of carcinoma cells. We conclude that administration of MJ with common chemotherapeutic drugs and glycolysis inhibitors bears a promise for effective anti-cancer therapy.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Cyclopentanes/pharmacology , Deoxyglucose/pharmacology , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Synergism , Mice , Oxylipins
15.
Chemotherapy ; 52(4): 205-9, 2006.
Article in English | MEDLINE | ID: mdl-16675903

ABSTRACT

The discovery of new antimalarial drugs is mandatory to improve the effectiveness of antimalarial prophylaxis and treatment. In this review, we focused on sphingolipids as potential new targets for antimalarial drugs. Inhibition of sphingomyelin and/or glucosylceramide synthases leads to increased intracellular concentrations of ceramide and results in growth inhibition of Plasmodium falciparum. In mammalian cells, ceramide mediates death by chemotherapeutic drugs. We demonstrated that ceramide mediates the antimalarial effect of artemisinin and mefloquine by depletion of glutathione levels. Furthermore, ceramide and artemisinin activated p38 mitogen-activated protein kinase in P. falciparum, thus inhibiting its growth, apparently by a non-apoptotic mechanism. In summary, we propose novel options of antimalarials based on ceramide cytotoxic activity.


Subject(s)
Antimalarials/therapeutic use , Ceramides/therapeutic use , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Sphingolipids/metabolism , Animals , Forecasting , Humans , Plasmodium falciparum/growth & development , Signal Transduction
16.
Anticancer Drugs ; 16(9): 911-6, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16162967

ABSTRACT

Since salicylate, a plant stress hormone, suppresses the growth of various types of cancer cells, it was deemed of interest to investigate whether the jasmonate family of plant stress hormones is endowed with anti-cancer activities. Cell lines representing a wide spectrum of malignancies, including prostate, breast and lung, exhibit sensitivity to the cytotoxic effects of methyl jasmonate (MJ). Jasmonates induced death in leukemic cells isolated from the blood of chronic lymphocytic leukemia (CLL) patients and increased significantly the survival of lymphoma-bearing mice. Among the naturally occurring jasmonates, MJ is the most active, while the synthetic methyl-4,5-didehydrojasmonate, was approximately 29-fold more active than MJ. The cytotoxic activity of MJ is independent of transcription and translation. Studies have suggested several mechanisms of action. It appears that while prolonged exposures to relatively low concentrations of jasmonates induce growth arrest and re-differentiation in myeloid leukemia cells, higher concentrations of MJ induce direct perturbation of cancer cell mitochondria, leading to the release of cytochrome c and eventual cell death. A most important characteristic of jasmonates is their ability to selectively kill cancer cells while sparing normal cells. Even within a mixed population of normal and leukemic cells derived from the blood of CLL patients, MJ killed preferentially the leukemic cells. In conclusion, jasmonates present a unique class of anti-cancer compounds which deserves continued research at the basic and pharmaceutical levels in order to yield novel chemotherapeutic agents against a range of neoplastic diseases.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclopentanes/pharmacology , Plant Growth Regulators/pharmacology , Acetates/pharmacology , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclopentanes/chemistry , Humans , Mitochondria/drug effects , Molecular Structure , Neoplasms/drug therapy , Oxylipins
17.
Br J Pharmacol ; 146(6): 800-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16170329

ABSTRACT

Mutations in p53, a tumor suppressor gene, occur in more than half of human cancers. Therefore, we tested the hypothesis that jasmonates (novel anticancer agents) can induce death in mutated p53-expressing cells. Two clones of B-lymphoma cells were studied, one expressing wild-type (wt) p53 and the other expressing mutated p53. Jasmonic acid and methyl jasmonate (0.25-3 mM) were each equally cytotoxic to both clones, whereas mutant p53-expressing cells were resistant to treatment with the radiomimetic agent neocarzinostatin and the chemotherapeutic agent bleomycin. Neocarzinostatin and bleomycin induced an elevation in the p53 levels in wt p53-expressing cells, whereas methyl jasmonate did not. Methyl jasmonate induced mostly apoptotic death in the wt p53-expressing cells, while no signs of early apoptosis were detected in mutant p53-expressing cells. In contrast, neocarzinostatin and bleomycin induced death only in wt p53-expressing cells, in an apoptotic mode. Methyl jasmonate induced a rapid depletion of ATP in both clones. In both clones, oligomycin (a mitochondrial ATP synthase inhibitor) did not increase ATP depletion induced by methyl jasmonate, whereas inhibition of glycolysis with 2-deoxyglucose did. High glucose levels protected both clones from methyl jasmonate-induced ATP depletion (and reduced methyl jasmonate-induced cytotoxicity), whereas high levels of pyruvate did not. These results suggest that methyl jasmonate induces ATP depletion mostly by compromising oxidative phosphorylation in the mitochondria. In conclusion, jasmonates can circumvent the resistance of mutant p53-expressing cells towards chemotherapy by inducing a nonapoptotic cell death.


Subject(s)
Acetates/pharmacology , Cyclopentanes/pharmacology , Mutation/genetics , Tumor Suppressor Protein p53/genetics , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Cell Survival/drug effects , Clone Cells , Deoxyglucose/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Humans , Immunoblotting , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Oligomycins/pharmacology , Oxylipins , Plant Growth Regulators/chemistry , Plant Growth Regulators/pharmacology , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism , Zinostatin/pharmacology
18.
Cancer Res ; 65(5): 1984-93, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15753398

ABSTRACT

We reported previously that jasmonates can kill human cancer cells. Many chemotherapeutic drugs induce mitochondrial membrane permeability transition, membrane depolarization, osmotic swelling, and release of cytochrome c, involving the opening of the permeability transition pore complex (PTPC). Because jasmonates exert their cytotoxic effects independent of transcription, translation, and p53 expression, we hypothesized that these compounds may act directly on mitochondria. Mitochondrial membrane depolarization was determined by flow cytometry, and cytochrome c release by Western blotting. Mitochondria were isolated by mechanical lysis and differential centrifugation. Cytotoxicity was measured by a tetrazolium-based assay, and mitochondrial swelling by spectrophotometry. Jasmonates induced membrane depolarization and cytochrome c release in intact human cancer cell lines. Jasmonates induced swelling in mitochondria isolated from Hep 3B hepatoma cells, but not in mitochondria isolated from 3T3 nontransformed cells or from normal lymphocytes, in a PTPC-mediated manner. Methyl jasmonate induced the release of cytochrome c from mitochondria isolated from cancer cell lines in a PTPC-mediated manner, but not from mitochondria isolated from normal lymphocytes. A correlation was found between cytotoxicity of methyl jasmonate and the percentage of leukemic cells in the blood of patients with chronic lymphocytic leukemia (CLL). Jasmonates induced membrane depolarization in CLL cells, and swelling and release of cytochrome c in mitochondria isolated from these cells. In conclusion, jasmonates act directly on mitochondria derived from cancer cells in a PTPC-mediated manner, and could therefore bypass premitochondrial apoptotic blocks. Jasmonates are promising candidates for the treatment of CLL and other types of cancer.


Subject(s)
Acetates/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cyclopentanes/pharmacology , Ion Channels/metabolism , Mitochondria/drug effects , Plant Growth Regulators/pharmacology , Animals , Blotting, Western , Calcium/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Membrane Permeability/drug effects , Cells, Cultured , Cytochromes c/metabolism , Fibroblasts/metabolism , Flow Cytometry , Humans , Ion Channels/antagonists & inhibitors , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lymphocytes/drug effects , Membrane Potentials/drug effects , Mice , Mitochondrial Membrane Transport Proteins , Mitochondrial Permeability Transition Pore , Mitochondrial Swelling , Oxylipins
19.
Br J Pharmacol ; 138(6): 1156-62, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12684272

ABSTRACT

1. Available anticancer drugs do not seem to modify the prognosis of metastatic melanoma. Salicylate and acetyl salicylic acid (aspirin) were found to suppress growth in a number of transformed cells, that is, prostate and colon. Therefore, we studied the direct effects of aspirin on metastatic B16 melanoma cells. 2. Aspirin at a plasma-attainable and nontoxic level suppressed the proliferation of B16 cells. 3. Aspirin induced the activation of p38 and c-Jun N-terminal kinase (JNK) mitogen-activated protein kinases. 4. Inhibition of JNK, but not p38, decreased the suppressive effect of aspirin upon the proliferation of B16 cells. 5. The aspirin-induced reduction in B16 proliferation was cumulative over time. 6. Aspirin and the chemotherapeutic drug 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) induced B16 cell death synergistically. 7. In addition to the murine B16 cell line, the proliferation of SK-28 human melanoma cells was also suppressed by aspirin. 8 In conclusion, aspirin suppresses the proliferation of metastatic B16 cells in a JNK-dependent mechanism.


Subject(s)
Aspirin/pharmacology , JNK Mitogen-Activated Protein Kinases , Melanoma, Experimental/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Anticarcinogenic Agents/metabolism , Cell Death/drug effects , Drug Synergism , Genes, jun/drug effects , Genes, jun/genetics , Humans , MAP Kinase Kinase 4 , Organophosphates/metabolism , Suppression, Genetic/drug effects , Suppression, Genetic/genetics , Tumor Cells, Cultured
20.
Cell Signal ; 14(1): 37-43, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11747987

ABSTRACT

P-glycoprotein (P-gp) mediates drug resistance. Protein kinase C (PKC) expression correlates with drug resistance in several types of cancer. We determined whether PKC signals the induction of P-gp in LNCaP human prostate cancer cells, and identified a specific isozyme involved, in a model of aspirin-induced P-glycoprotein expression. An inhibitor of PKC activity, and a specific peptide inhibitor of PKC epsilon translocation, suppressed the induction of P-gp. The PKC activator ingenol, but not OAG, induced P-gp expression in a dose-dependent manner. Based on our results, we conclude that PKC epsilon mediates the induction of P-gp. Accordingly, PKC epsilon is activated and translocates from the membrane fraction to the cytoskeleton fraction in aspirin-treated cells. The findings of this study point to PKC epsilon as a signalling molecule for the induction of P-gp in LNCaP prostate cancer cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Carcinoma/metabolism , Isoenzymes/physiology , Prostatic Neoplasms/metabolism , Protein Kinase C-epsilon/physiology , Aspirin/pharmacology , Carcinoma/enzymology , Diterpenes/pharmacology , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , Indoles/pharmacology , Isoenzymes/antagonists & inhibitors , Kinetics , Male , Peptide Fragments/pharmacology , Prostatic Neoplasms/enzymology , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Transport/drug effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...