Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 4: e664, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23764845

ABSTRACT

The anti-tumoral effects of cannabinoids have been described in different tumor systems, including pancreatic adenocarcinoma, but their mechanism of action remains unclear. We used cannabinoids specific for the CB1 (ACPA) and CB2 (GW) receptors and metabolomic analyses to unravel the potential pathways mediating cannabinoid-dependent inhibition of pancreatic cancer cell growth. Panc1 cells treated with cannabinoids show elevated AMPK activation induced by a ROS-dependent increase of AMP/ATP ratio. ROS promote nuclear translocation of GAPDH, which is further amplified by AMPK, thereby attenuating glycolysis. Furthermore, ROS determine the accumulation of NADH, suggestive of a blockage in the respiratory chain, which in turn inhibits the Krebs cycle. Concomitantly, inhibition of Akt/c-Myc pathway leads to decreased activity of both the pyruvate kinase isoform M2 (PKM2), further downregulating glycolysis, and glutamine uptake. Altogether, these alterations of pancreatic cancer cell metabolism mediated by cannabinoids result in a strong induction of autophagy and in the inhibition of cell growth.


Subject(s)
Adenylate Kinase/metabolism , Antineoplastic Agents/pharmacology , Arachidonic Acids/pharmacology , Autophagy , Cannabinoids/pharmacology , Indoles/pharmacology , Morpholines/pharmacology , Acetylcysteine/pharmacology , Adenocarcinoma , Adenosine Monophosphate/metabolism , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Cell Nucleus/enzymology , Citric Acid Cycle/drug effects , Drug Screening Assays, Antitumor , Energy Metabolism , Enzyme Activation , Glutamine/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Glycolysis/drug effects , Humans , Pancreatic Neoplasms , Reactive Oxygen Species/metabolism , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB2/agonists
2.
Cell Death Dis ; 2: e152, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21525939

ABSTRACT

Gemcitabine (GEM, 2',2'-difluorodeoxycytidine) is currently used in advanced pancreatic adenocarcinoma, with a response rate of < 20%. The purpose of our work was to improve GEM activity by addition of cannabinoids. Here, we show that GEM induces both cannabinoid receptor-1 (CB1) and cannabinoid receptor-2 (CB2) receptors by an NF-κB-dependent mechanism and that its association with cannabinoids synergistically inhibits pancreatic adenocarcinoma cell growth and increases reactive oxygen species (ROS) induced by single treatments. The antiproliferative synergism is prevented by the radical scavenger N-acetyl-L-cysteine and by the specific NF-κB inhibitor BAY 11-7085, demonstrating that the induction of ROS by GEM/cannabinoids and of NF-κB by GEM is required for this effect. In addition, we report that neither apoptotic nor cytostatic mechanisms are responsible for the synergistic cell growth inhibition, which is strictly associated with the enhancement of endoplasmic reticulum stress and autophagic cell death. Noteworthy, the antiproliferative synergism is stronger in GEM-resistant pancreatic cancer cell lines compared with GEM-sensitive pancreatic cancer cell lines. The combined treatment strongly inhibits growth of human pancreatic tumor cells xenografted in nude mice without apparent toxic effects. These findings support a key role of the ROS-dependent activation of an autophagic program in the synergistic growth inhibition induced by GEM/cannabinoid combination in human pancreatic cancer cells.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Autophagy/drug effects , Pancreatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Animals , Cannabinoids/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/drug therapy , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, Cannabinoid, CB2/metabolism , Stress, Physiological , Transcription, Genetic/drug effects , Transplantation, Heterologous , Gemcitabine
3.
Biochim Biophys Acta ; 1793(2): 273-80, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18951928

ABSTRACT

We show that treatment with non-toxic doses of zinc in association to the ionophore compound pyrrolidine dithiocarbamate (PDTC) inhibits p53(-/-) pancreatic cancer cell growth much more efficiently than gemcitabine, the gold standard chemotherapeutic agent for pancreatic cancer. Both the metal chelator N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine and the radical scavenger N-acetyl-l-cysteine are able to recover cell growth inhibition by Zn/PDTC, demonstrating that this effect depends on the increased levels of intracellular zinc and of reactive oxygen species (ROS). Zn/PDTC treatment induces a strong apoptotic cell death that is associated to ROS-dependent nuclear translocation of the mitochondrial factor AIF, but not to the regulation of apoptotic genes and caspase activation. Primary fibroblasts are more resistant than pancreatic cancer cells to Zn/PDTC treatment and exhibit a lower basal and Zn/PDTC-induced enhancement of intracellular zinc. We show that Zn/PDTC induces p53 proteasomal degradation and that the proteasome inhibitor MG132 further increases fibroblast growth inhibition by Zn/PDTC, suggesting that p53 degradation plays an important role in fibroblast resistance to Zn/PDTC.


Subject(s)
Adenocarcinoma/pathology , Apoptosis Inducing Factor/metabolism , Apoptosis , Pancreatic Neoplasms/pathology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/deficiency , Zinc/metabolism , Adenocarcinoma/metabolism , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Ethylenediamines/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Leupeptins/pharmacology , Mitochondria/drug effects , Mitochondria/enzymology , Models, Biological , Pancreatic Neoplasms/metabolism , Protein Transport/drug effects , Pyrrolidines/pharmacology , Thiocarbamates/pharmacology , Tumor Suppressor Protein p53/genetics
4.
J Cell Biochem ; 104(1): 202-12, 2008 May 01.
Article in English | MEDLINE | ID: mdl-17979179

ABSTRACT

We investigated the ability of the zinc chelator N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine (TPEN) to reduce pancreatic cancer cell viability. TPEN was much more efficient to inhibit pancreatic adenocarcinoma cell growth than a panel of anti-cancer drugs, including 5-fluorouracil, irinotecan, cisplatin, edelfosine, trichostatin A, mitomycin C, and gemcitabine, the gold standard chemotherapeutic agent for pancreatic cancer. Moreover, TPEN showed a dose- and time-dependent anti-proliferative effect significantly higher on pancreatic cancer cells than on normal primary fibroblasts. This effect may be explained by a significantly higher zinc depletion by TPEN in pancreatic cancer cells as compared to fibroblasts. Cell viability reduction by TPEN was associated to both G1-phase cell cycle arrest and apoptosis, and to the increased ratio of the expression level of cyclin-Cdk inhibitor versus cyclin genes and apoptotic versus anti-apoptotic genes. Finally, we show that apoptotic cell death induced by TPEN involved mitochondrial injury and caspase 3 and caspase 8 activation. In this study, we suggest that zinc depletion may be an efficient strategy in the treatment of pancreatic cancer because of its reduced antiproliferative effect on normal cells.


Subject(s)
Cell Proliferation/drug effects , Ethylenediamines/pharmacology , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms/therapy , Zinc/deficiency , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Chelating Agents/pharmacology , Chelating Agents/therapeutic use , Ethylenediamines/therapeutic use , Humans , Pancreatic Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...